WO2013006479A2 - Mammalian cell culture - Google Patents
Mammalian cell culture Download PDFInfo
- Publication number
- WO2013006479A2 WO2013006479A2 PCT/US2012/045070 US2012045070W WO2013006479A2 WO 2013006479 A2 WO2013006479 A2 WO 2013006479A2 US 2012045070 W US2012045070 W US 2012045070W WO 2013006479 A2 WO2013006479 A2 WO 2013006479A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- perfusion
- asparagine
- culture
- cell
- serum
- Prior art date
Links
- 238000004113 cell culture Methods 0.000 title claims abstract description 95
- 210000004962 mammalian cell Anatomy 0.000 title claims abstract description 60
- 238000000034 method Methods 0.000 claims abstract description 124
- 230000010261 cell growth Effects 0.000 claims abstract description 14
- 238000012258 culturing Methods 0.000 claims abstract description 4
- 210000004027 cell Anatomy 0.000 claims description 296
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 claims description 246
- 230000010412 perfusion Effects 0.000 claims description 186
- 229960001230 asparagine Drugs 0.000 claims description 131
- 238000004519 manufacturing process Methods 0.000 claims description 95
- 239000002609 medium Substances 0.000 claims description 68
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 claims description 43
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 claims description 43
- 230000003698 anagen phase Effects 0.000 claims description 22
- 230000001965 increasing effect Effects 0.000 claims description 21
- 239000012526 feed medium Substances 0.000 claims description 18
- 239000006143 cell culture medium Substances 0.000 claims description 15
- 235000003642 hunger Nutrition 0.000 claims description 15
- 239000004017 serum-free culture medium Substances 0.000 claims description 15
- 230000037351 starvation Effects 0.000 claims description 15
- 239000001963 growth medium Substances 0.000 claims description 14
- 230000001939 inductive effect Effects 0.000 claims description 14
- 230000014616 translation Effects 0.000 claims description 12
- 238000009472 formulation Methods 0.000 claims description 10
- 238000003306 harvesting Methods 0.000 claims description 10
- 239000000203 mixture Substances 0.000 claims description 10
- 230000000670 limiting effect Effects 0.000 claims description 9
- 230000007704 transition Effects 0.000 claims description 8
- 102000004127 Cytokines Human genes 0.000 claims description 7
- 108090000695 Cytokines Proteins 0.000 claims description 7
- 230000006698 induction Effects 0.000 claims description 5
- 241000699802 Cricetulus griseus Species 0.000 claims description 3
- 210000001672 ovary Anatomy 0.000 claims description 3
- 230000001502 supplementing effect Effects 0.000 claims description 2
- 108090000623 proteins and genes Proteins 0.000 description 43
- 102000004169 proteins and genes Human genes 0.000 description 40
- 235000018102 proteins Nutrition 0.000 description 39
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 30
- 235000009582 asparagine Nutrition 0.000 description 30
- 230000035899 viability Effects 0.000 description 22
- 230000012010 growth Effects 0.000 description 19
- 108090000765 processed proteins & peptides Proteins 0.000 description 17
- 239000000047 product Substances 0.000 description 16
- 102000004196 processed proteins & peptides Human genes 0.000 description 15
- 239000000427 antigen Substances 0.000 description 14
- 102000036639 antigens Human genes 0.000 description 14
- 108091007433 antigens Proteins 0.000 description 14
- 230000035611 feeding Effects 0.000 description 14
- 239000007787 solid Substances 0.000 description 14
- 229920001184 polypeptide Polymers 0.000 description 13
- 230000008569 process Effects 0.000 description 11
- 102000005962 receptors Human genes 0.000 description 11
- 108020003175 receptors Proteins 0.000 description 11
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 10
- 239000003446 ligand Substances 0.000 description 10
- 238000011020 pilot scale process Methods 0.000 description 9
- 229930182816 L-glutamine Natural products 0.000 description 8
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 8
- 102100040247 Tumor necrosis factor Human genes 0.000 description 8
- 238000013019 agitation Methods 0.000 description 8
- 238000011021 bench scale process Methods 0.000 description 8
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 7
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 7
- 230000002354 daily effect Effects 0.000 description 7
- 239000008103 glucose Substances 0.000 description 7
- 206010028980 Neoplasm Diseases 0.000 description 6
- 150000001413 amino acids Chemical class 0.000 description 6
- 239000010432 diamond Substances 0.000 description 6
- 238000002474 experimental method Methods 0.000 description 6
- 238000001914 filtration Methods 0.000 description 6
- 235000015097 nutrients Nutrition 0.000 description 6
- 210000004102 animal cell Anatomy 0.000 description 5
- 239000005557 antagonist Substances 0.000 description 5
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 5
- 230000007423 decrease Effects 0.000 description 5
- 239000012510 hollow fiber Substances 0.000 description 5
- 229910052760 oxygen Inorganic materials 0.000 description 5
- 239000001301 oxygen Substances 0.000 description 5
- QGZKDVFQNNGYKY-UHFFFAOYSA-N Ammonia Chemical compound N QGZKDVFQNNGYKY-UHFFFAOYSA-N 0.000 description 4
- 239000002028 Biomass Substances 0.000 description 4
- 238000010923 batch production Methods 0.000 description 4
- 230000027455 binding Effects 0.000 description 4
- 230000003833 cell viability Effects 0.000 description 4
- 238000009295 crossflow filtration Methods 0.000 description 4
- 238000012136 culture method Methods 0.000 description 4
- 239000003814 drug Substances 0.000 description 4
- 230000000694 effects Effects 0.000 description 4
- 238000004128 high performance liquid chromatography Methods 0.000 description 4
- 230000006872 improvement Effects 0.000 description 4
- 230000000977 initiatory effect Effects 0.000 description 4
- 238000011081 inoculation Methods 0.000 description 4
- 230000002503 metabolic effect Effects 0.000 description 4
- 101001031613 Homo sapiens Fibroleukin Proteins 0.000 description 3
- 108060003951 Immunoglobulin Proteins 0.000 description 3
- 108010002350 Interleukin-2 Proteins 0.000 description 3
- 102000000588 Interleukin-2 Human genes 0.000 description 3
- 102000014128 RANK Ligand Human genes 0.000 description 3
- 108010025832 RANK Ligand Proteins 0.000 description 3
- 108010022394 Threonine synthase Proteins 0.000 description 3
- 108060008683 Tumor Necrosis Factor Receptor Proteins 0.000 description 3
- 238000009825 accumulation Methods 0.000 description 3
- 229940024606 amino acid Drugs 0.000 description 3
- 235000001014 amino acid Nutrition 0.000 description 3
- 239000000872 buffer Substances 0.000 description 3
- 239000006227 byproduct Substances 0.000 description 3
- 230000001276 controlling effect Effects 0.000 description 3
- 229910003460 diamond Inorganic materials 0.000 description 3
- 102000004419 dihydrofolate reductase Human genes 0.000 description 3
- 238000011143 downstream manufacturing Methods 0.000 description 3
- 238000005516 engineering process Methods 0.000 description 3
- 239000012634 fragment Substances 0.000 description 3
- 230000014509 gene expression Effects 0.000 description 3
- 239000003102 growth factor Substances 0.000 description 3
- 102000018358 immunoglobulin Human genes 0.000 description 3
- 239000000411 inducer Substances 0.000 description 3
- 229940047124 interferons Drugs 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 210000002966 serum Anatomy 0.000 description 3
- 102000003298 tumor necrosis factor receptor Human genes 0.000 description 3
- 102100038080 B-cell receptor CD22 Human genes 0.000 description 2
- 101150013553 CD40 gene Proteins 0.000 description 2
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 2
- 108010022366 Carcinoembryonic Antigen Proteins 0.000 description 2
- 102100025475 Carcinoembryonic antigen-related cell adhesion molecule 5 Human genes 0.000 description 2
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 2
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- 108010066687 Epithelial Cell Adhesion Molecule Proteins 0.000 description 2
- 102000018651 Epithelial Cell Adhesion Molecule Human genes 0.000 description 2
- 102100038647 Fibroleukin Human genes 0.000 description 2
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 2
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 2
- 102000006354 HLA-DR Antigens Human genes 0.000 description 2
- 108010058597 HLA-DR Antigens Proteins 0.000 description 2
- 101000884305 Homo sapiens B-cell receptor CD22 Proteins 0.000 description 2
- 101001133056 Homo sapiens Mucin-1 Proteins 0.000 description 2
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 2
- 241000725303 Human immunodeficiency virus Species 0.000 description 2
- 108010064600 Intercellular Adhesion Molecule-3 Proteins 0.000 description 2
- 102100037871 Intercellular adhesion molecule 3 Human genes 0.000 description 2
- 108010074328 Interferon-gamma Proteins 0.000 description 2
- 102000008070 Interferon-gamma Human genes 0.000 description 2
- 102000014150 Interferons Human genes 0.000 description 2
- 108010050904 Interferons Proteins 0.000 description 2
- 102000004557 Interleukin-18 Receptors Human genes 0.000 description 2
- 108010017537 Interleukin-18 Receptors Proteins 0.000 description 2
- 102000010789 Interleukin-2 Receptors Human genes 0.000 description 2
- 108010038453 Interleukin-2 Receptors Proteins 0.000 description 2
- 102100026878 Interleukin-2 receptor subunit alpha Human genes 0.000 description 2
- 108010002386 Interleukin-3 Proteins 0.000 description 2
- 102100039064 Interleukin-3 Human genes 0.000 description 2
- 102000010787 Interleukin-4 Receptors Human genes 0.000 description 2
- 108010038486 Interleukin-4 Receptors Proteins 0.000 description 2
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 2
- 102000004058 Leukemia inhibitory factor Human genes 0.000 description 2
- 108090000581 Leukemia inhibitory factor Proteins 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- 102100034256 Mucin-1 Human genes 0.000 description 2
- BNQSTAOJRULKNX-UHFFFAOYSA-N N-(6-acetamidohexyl)acetamide Chemical compound CC(=O)NCCCCCCNC(C)=O BNQSTAOJRULKNX-UHFFFAOYSA-N 0.000 description 2
- 102000004140 Oncostatin M Human genes 0.000 description 2
- 108090000630 Oncostatin M Proteins 0.000 description 2
- 102000003982 Parathyroid hormone Human genes 0.000 description 2
- 108090000445 Parathyroid hormone Proteins 0.000 description 2
- 108010038036 Receptor Activator of Nuclear Factor-kappa B Proteins 0.000 description 2
- 102000010498 Receptor Activator of Nuclear Factor-kappa B Human genes 0.000 description 2
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 2
- 108010000449 TNF-Related Apoptosis-Inducing Ligand Receptors Proteins 0.000 description 2
- 102000002259 TNF-Related Apoptosis-Inducing Ligand Receptors Human genes 0.000 description 2
- 102100036922 Tumor necrosis factor ligand superfamily member 13B Human genes 0.000 description 2
- 102100040245 Tumor necrosis factor receptor superfamily member 5 Human genes 0.000 description 2
- -1 WO 01/36637) Proteins 0.000 description 2
- 102000013529 alpha-Fetoproteins Human genes 0.000 description 2
- 108010026331 alpha-Fetoproteins Proteins 0.000 description 2
- 229910021529 ammonia Inorganic materials 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- RYYVLZVUVIJVGH-UHFFFAOYSA-N caffeine Chemical compound CN1C(=O)N(C)C(=O)C2=C1N=CN2C RYYVLZVUVIJVGH-UHFFFAOYSA-N 0.000 description 2
- 230000022131 cell cycle Effects 0.000 description 2
- 230000004663 cell proliferation Effects 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 210000001072 colon Anatomy 0.000 description 2
- 238000010960 commercial process Methods 0.000 description 2
- 230000003247 decreasing effect Effects 0.000 description 2
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 2
- 230000036541 health Effects 0.000 description 2
- 230000001976 improved effect Effects 0.000 description 2
- 239000003112 inhibitor Substances 0.000 description 2
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 2
- 238000012007 large scale cell culture Methods 0.000 description 2
- 150000002632 lipids Chemical class 0.000 description 2
- 239000012092 media component Substances 0.000 description 2
- 238000010369 molecular cloning Methods 0.000 description 2
- 238000005457 optimization Methods 0.000 description 2
- 229960001319 parathyroid hormone Drugs 0.000 description 2
- 239000000199 parathyroid hormone Substances 0.000 description 2
- 238000000746 purification Methods 0.000 description 2
- 230000000717 retained effect Effects 0.000 description 2
- 150000003839 salts Chemical class 0.000 description 2
- 238000004062 sedimentation Methods 0.000 description 2
- 235000002639 sodium chloride Nutrition 0.000 description 2
- 238000003860 storage Methods 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 230000000153 supplemental effect Effects 0.000 description 2
- 230000002459 sustained effect Effects 0.000 description 2
- 235000013343 vitamin Nutrition 0.000 description 2
- 229940088594 vitamin Drugs 0.000 description 2
- 239000011782 vitamin Substances 0.000 description 2
- 229930003231 vitamin Natural products 0.000 description 2
- RTQWWZBSTRGEAV-PKHIMPSTSA-N 2-[[(2s)-2-[bis(carboxymethyl)amino]-3-[4-(methylcarbamoylamino)phenyl]propyl]-[2-[bis(carboxymethyl)amino]propyl]amino]acetic acid Chemical compound CNC(=O)NC1=CC=C(C[C@@H](CN(CC(C)N(CC(O)=O)CC(O)=O)CC(O)=O)N(CC(O)=O)CC(O)=O)C=C1 RTQWWZBSTRGEAV-PKHIMPSTSA-N 0.000 description 1
- MJZJYWCQPMNPRM-UHFFFAOYSA-N 6,6-dimethyl-1-[3-(2,4,5-trichlorophenoxy)propoxy]-1,6-dihydro-1,3,5-triazine-2,4-diamine Chemical compound CC1(C)N=C(N)N=C(N)N1OCCCOC1=CC(Cl)=C(Cl)C=C1Cl MJZJYWCQPMNPRM-UHFFFAOYSA-N 0.000 description 1
- 108091007505 ADAM17 Proteins 0.000 description 1
- 230000005730 ADP ribosylation Effects 0.000 description 1
- 108010054404 Adenylyl-sulfate kinase Proteins 0.000 description 1
- 102100034608 Angiopoietin-2 Human genes 0.000 description 1
- 108010048036 Angiopoietin-2 Proteins 0.000 description 1
- 102000005666 Apolipoprotein A-I Human genes 0.000 description 1
- 108010059886 Apolipoprotein A-I Proteins 0.000 description 1
- 102100029470 Apolipoprotein E Human genes 0.000 description 1
- 101710095339 Apolipoprotein E Proteins 0.000 description 1
- 108010070255 Aspartate-ammonia ligase Proteins 0.000 description 1
- 108010028006 B-Cell Activating Factor Proteins 0.000 description 1
- 102100022005 B-lymphocyte antigen CD20 Human genes 0.000 description 1
- 108010039209 Blood Coagulation Factors Proteins 0.000 description 1
- 102000015081 Blood Coagulation Factors Human genes 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 102000004219 Brain-derived neurotrophic factor Human genes 0.000 description 1
- 108090000715 Brain-derived neurotrophic factor Proteins 0.000 description 1
- FERIUCNNQQJTOY-UHFFFAOYSA-M Butyrate Chemical compound CCCC([O-])=O FERIUCNNQQJTOY-UHFFFAOYSA-M 0.000 description 1
- FERIUCNNQQJTOY-UHFFFAOYSA-N Butyric acid Natural products CCCC(O)=O FERIUCNNQQJTOY-UHFFFAOYSA-N 0.000 description 1
- 101710167766 C-type lectin domain family 11 member A Proteins 0.000 description 1
- 102100032528 C-type lectin domain family 11 member A Human genes 0.000 description 1
- 108010008629 CA-125 Antigen Proteins 0.000 description 1
- 102000007269 CA-125 Antigen Human genes 0.000 description 1
- 102100024217 CAMPATH-1 antigen Human genes 0.000 description 1
- 102000007499 CD27 Ligand Human genes 0.000 description 1
- 108010046080 CD27 Ligand Proteins 0.000 description 1
- 102100027207 CD27 antigen Human genes 0.000 description 1
- 102000004634 CD30 Ligand Human genes 0.000 description 1
- 108010017987 CD30 Ligand Proteins 0.000 description 1
- 102100032912 CD44 antigen Human genes 0.000 description 1
- 108010065524 CD52 Antigen Proteins 0.000 description 1
- 102000055006 Calcitonin Human genes 0.000 description 1
- 108060001064 Calcitonin Proteins 0.000 description 1
- 102000013602 Cardiac Myosins Human genes 0.000 description 1
- 108010051609 Cardiac Myosins Proteins 0.000 description 1
- 108010055166 Chemokine CCL5 Proteins 0.000 description 1
- 102000001327 Chemokine CCL5 Human genes 0.000 description 1
- 102100022641 Coagulation factor IX Human genes 0.000 description 1
- 206010009944 Colon cancer Diseases 0.000 description 1
- 108010071942 Colony-Stimulating Factors Proteins 0.000 description 1
- 102000007644 Colony-Stimulating Factors Human genes 0.000 description 1
- 108010028773 Complement C5 Proteins 0.000 description 1
- 101000774560 Crotalus atrox Zinc metalloproteinase-disintegrin-like VAP1 Proteins 0.000 description 1
- 102100039498 Cytotoxic T-lymphocyte protein 4 Human genes 0.000 description 1
- 102000010170 Death domains Human genes 0.000 description 1
- 108050001718 Death domains Proteins 0.000 description 1
- 101800001224 Disintegrin Proteins 0.000 description 1
- 102100031111 Disintegrin and metalloproteinase domain-containing protein 17 Human genes 0.000 description 1
- 102000001301 EGF receptor Human genes 0.000 description 1
- 108060006698 EGF receptor Proteins 0.000 description 1
- 102100029722 Ectonucleoside triphosphate diphosphohydrolase 1 Human genes 0.000 description 1
- 101800003838 Epidermal growth factor Proteins 0.000 description 1
- 108010008165 Etanercept Proteins 0.000 description 1
- 108010076282 Factor IX Proteins 0.000 description 1
- 108010054218 Factor VIII Proteins 0.000 description 1
- 102000001690 Factor VIII Human genes 0.000 description 1
- 102000009109 Fc receptors Human genes 0.000 description 1
- 108010087819 Fc receptors Proteins 0.000 description 1
- 102000003972 Fibroblast growth factor 7 Human genes 0.000 description 1
- 108090000385 Fibroblast growth factor 7 Proteins 0.000 description 1
- 230000005526 G1 to G0 transition Effects 0.000 description 1
- 208000032612 Glial tumor Diseases 0.000 description 1
- 206010018338 Glioma Diseases 0.000 description 1
- 102400000321 Glucagon Human genes 0.000 description 1
- 108060003199 Glucagon Proteins 0.000 description 1
- 108010017544 Glucosylceramidase Proteins 0.000 description 1
- 102000004547 Glucosylceramidase Human genes 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 description 1
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 description 1
- 108010054017 Granulocyte Colony-Stimulating Factor Receptors Proteins 0.000 description 1
- 102100039622 Granulocyte colony-stimulating factor receptor Human genes 0.000 description 1
- 102000016355 Granulocyte-Macrophage Colony-Stimulating Factor Receptors Human genes 0.000 description 1
- 108010092372 Granulocyte-Macrophage Colony-Stimulating Factor Receptors Proteins 0.000 description 1
- 108010051696 Growth Hormone Proteins 0.000 description 1
- 241000700721 Hepatitis B virus Species 0.000 description 1
- 108090000100 Hepatocyte Growth Factor Proteins 0.000 description 1
- 102100021866 Hepatocyte growth factor Human genes 0.000 description 1
- 101000897405 Homo sapiens B-lymphocyte antigen CD20 Proteins 0.000 description 1
- 101000914511 Homo sapiens CD27 antigen Proteins 0.000 description 1
- 101000868273 Homo sapiens CD44 antigen Proteins 0.000 description 1
- 101000889276 Homo sapiens Cytotoxic T-lymphocyte protein 4 Proteins 0.000 description 1
- 101001012447 Homo sapiens Ectonucleoside triphosphate diphosphohydrolase 1 Proteins 0.000 description 1
- 101000935040 Homo sapiens Integrin beta-2 Proteins 0.000 description 1
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 1
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 1
- 101000878605 Homo sapiens Low affinity immunoglobulin epsilon Fc receptor Proteins 0.000 description 1
- 101000946889 Homo sapiens Monocyte differentiation antigen CD14 Proteins 0.000 description 1
- 101000934338 Homo sapiens Myeloid cell surface antigen CD33 Proteins 0.000 description 1
- 101001012157 Homo sapiens Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 description 1
- 101000851376 Homo sapiens Tumor necrosis factor receptor superfamily member 8 Proteins 0.000 description 1
- 102100023915 Insulin Human genes 0.000 description 1
- 108090001061 Insulin Proteins 0.000 description 1
- 108090000723 Insulin-Like Growth Factor I Proteins 0.000 description 1
- 108010008212 Integrin alpha4beta1 Proteins 0.000 description 1
- 102100025390 Integrin beta-2 Human genes 0.000 description 1
- 102000000589 Interleukin-1 Human genes 0.000 description 1
- 108010002352 Interleukin-1 Proteins 0.000 description 1
- 102000019223 Interleukin-1 receptor Human genes 0.000 description 1
- 108050006617 Interleukin-1 receptor Proteins 0.000 description 1
- 102000003814 Interleukin-10 Human genes 0.000 description 1
- 108090000174 Interleukin-10 Proteins 0.000 description 1
- 102000004559 Interleukin-13 Receptors Human genes 0.000 description 1
- 108010017511 Interleukin-13 Receptors Proteins 0.000 description 1
- 102000004556 Interleukin-15 Receptors Human genes 0.000 description 1
- 108010017535 Interleukin-15 Receptors Proteins 0.000 description 1
- 102000004554 Interleukin-17 Receptors Human genes 0.000 description 1
- 108010017525 Interleukin-17 Receptors Proteins 0.000 description 1
- 102000004388 Interleukin-4 Human genes 0.000 description 1
- 108090000978 Interleukin-4 Proteins 0.000 description 1
- 108010002616 Interleukin-5 Proteins 0.000 description 1
- 102000000743 Interleukin-5 Human genes 0.000 description 1
- 102000010781 Interleukin-6 Receptors Human genes 0.000 description 1
- 108010038501 Interleukin-6 Receptors Proteins 0.000 description 1
- 108010002586 Interleukin-7 Proteins 0.000 description 1
- 102000000704 Interleukin-7 Human genes 0.000 description 1
- 108090001007 Interleukin-8 Proteins 0.000 description 1
- 102000004890 Interleukin-8 Human genes 0.000 description 1
- 102000015696 Interleukins Human genes 0.000 description 1
- 108010063738 Interleukins Proteins 0.000 description 1
- 239000007760 Iscove's Modified Dulbecco's Medium Substances 0.000 description 1
- LPHGQDQBBGAPDZ-UHFFFAOYSA-N Isocaffeine Natural products CN1C(=O)N(C)C(=O)C2=C1N(C)C=N2 LPHGQDQBBGAPDZ-UHFFFAOYSA-N 0.000 description 1
- 208000008839 Kidney Neoplasms Diseases 0.000 description 1
- 108010092694 L-Selectin Proteins 0.000 description 1
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 1
- 102000016551 L-selectin Human genes 0.000 description 1
- JVTAAEKCZFNVCJ-UHFFFAOYSA-M Lactate Chemical compound CC(O)C([O-])=O JVTAAEKCZFNVCJ-UHFFFAOYSA-M 0.000 description 1
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 1
- 102100038007 Low affinity immunoglobulin epsilon Fc receptor Human genes 0.000 description 1
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 1
- 102000003959 Lymphotoxin-beta Human genes 0.000 description 1
- 108090000362 Lymphotoxin-beta Proteins 0.000 description 1
- 102000005741 Metalloproteases Human genes 0.000 description 1
- 108010006035 Metalloproteases Proteins 0.000 description 1
- 102100035877 Monocyte differentiation antigen CD14 Human genes 0.000 description 1
- 101000845218 Mus musculus Thymic stromal lymphopoietin Proteins 0.000 description 1
- 102100025243 Myeloid cell surface antigen CD33 Human genes 0.000 description 1
- 108010025020 Nerve Growth Factor Proteins 0.000 description 1
- 102000015336 Nerve Growth Factor Human genes 0.000 description 1
- 206010029260 Neuroblastoma Diseases 0.000 description 1
- 108090000742 Neurotrophin 3 Proteins 0.000 description 1
- 102100029268 Neurotrophin-3 Human genes 0.000 description 1
- 102000004473 OX40 Ligand Human genes 0.000 description 1
- 108010042215 OX40 Ligand Proteins 0.000 description 1
- 108010035042 Osteoprotegerin Proteins 0.000 description 1
- 206010061902 Pancreatic neoplasm Diseases 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 102000015795 Platelet Membrane Glycoproteins Human genes 0.000 description 1
- 108010010336 Platelet Membrane Glycoproteins Proteins 0.000 description 1
- 102100033237 Pro-epidermal growth factor Human genes 0.000 description 1
- 102000001253 Protein Kinase Human genes 0.000 description 1
- 239000012980 RPMI-1640 medium Substances 0.000 description 1
- 102100030086 Receptor tyrosine-protein kinase erbB-2 Human genes 0.000 description 1
- 206010038389 Renal cancer Diseases 0.000 description 1
- 108050006698 Sclerostin Proteins 0.000 description 1
- 102100034201 Sclerostin Human genes 0.000 description 1
- 102000013275 Somatomedins Human genes 0.000 description 1
- 102100038803 Somatotropin Human genes 0.000 description 1
- 102100039024 Sphingosine kinase 1 Human genes 0.000 description 1
- 108010039445 Stem Cell Factor Proteins 0.000 description 1
- 241000194019 Streptococcus mutans Species 0.000 description 1
- 102000019197 Superoxide Dismutase Human genes 0.000 description 1
- 108010012715 Superoxide dismutase Proteins 0.000 description 1
- 102000003978 Tissue Plasminogen Activator Human genes 0.000 description 1
- 108090000373 Tissue Plasminogen Activator Proteins 0.000 description 1
- 102100032236 Tumor necrosis factor receptor superfamily member 11B Human genes 0.000 description 1
- 101710165473 Tumor necrosis factor receptor superfamily member 4 Proteins 0.000 description 1
- 102100022153 Tumor necrosis factor receptor superfamily member 4 Human genes 0.000 description 1
- 102100036857 Tumor necrosis factor receptor superfamily member 8 Human genes 0.000 description 1
- 108091008605 VEGF receptors Proteins 0.000 description 1
- 102000009484 Vascular Endothelial Growth Factor Receptors Human genes 0.000 description 1
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 description 1
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- 229960000446 abciximab Drugs 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 229960002964 adalimumab Drugs 0.000 description 1
- 238000004115 adherent culture Methods 0.000 description 1
- 230000001464 adherent effect Effects 0.000 description 1
- 229960000548 alemtuzumab Drugs 0.000 description 1
- 102000015395 alpha 1-Antitrypsin Human genes 0.000 description 1
- 108010050122 alpha 1-Antitrypsin Proteins 0.000 description 1
- 229940024142 alpha 1-antitrypsin Drugs 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 125000000539 amino acid group Chemical group 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 239000002518 antifoaming agent Substances 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 229950001863 bapineuzumab Drugs 0.000 description 1
- 229960004669 basiliximab Drugs 0.000 description 1
- 229960005347 belatacept Drugs 0.000 description 1
- 229960003270 belimumab Drugs 0.000 description 1
- 229960000397 bevacizumab Drugs 0.000 description 1
- 230000002051 biphasic effect Effects 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 239000003114 blood coagulation factor Substances 0.000 description 1
- 229940077737 brain-derived neurotrophic factor Drugs 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- 229960002874 briakinumab Drugs 0.000 description 1
- 229960001948 caffeine Drugs 0.000 description 1
- VJEONQKOZGKCAK-UHFFFAOYSA-N caffeine Natural products CN1C(=O)N(C)C(=O)C2=C1C=CN2C VJEONQKOZGKCAK-UHFFFAOYSA-N 0.000 description 1
- BBBFJLBPOGFECG-VJVYQDLKSA-N calcitonin Chemical compound N([C@H](C(=O)N[C@@H](CC(C)C)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=1NC=NC=1)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)NCC(=O)N[C@@H](CO)C(=O)NCC(=O)N[C@@H]([C@@H](C)O)C(=O)N1[C@@H](CCC1)C(N)=O)C(C)C)C(=O)[C@@H]1CSSC[C@H](N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CO)C(=O)N[C@@H]([C@@H](C)O)C(=O)N1 BBBFJLBPOGFECG-VJVYQDLKSA-N 0.000 description 1
- 229960004015 calcitonin Drugs 0.000 description 1
- 229960001838 canakinumab Drugs 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 229940077731 carbohydrate nutrients Drugs 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 229910002092 carbon dioxide Inorganic materials 0.000 description 1
- 239000001569 carbon dioxide Substances 0.000 description 1
- 230000025084 cell cycle arrest Effects 0.000 description 1
- 230000011712 cell development Effects 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 229960003115 certolizumab pegol Drugs 0.000 description 1
- 229960005395 cetuximab Drugs 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 208000029742 colonic neoplasm Diseases 0.000 description 1
- 229940047120 colony stimulating factors Drugs 0.000 description 1
- 238000012777 commercial manufacturing Methods 0.000 description 1
- 229950007276 conatumumab Drugs 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 210000004748 cultured cell Anatomy 0.000 description 1
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 229960001251 denosumab Drugs 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 229940079593 drug Drugs 0.000 description 1
- 229960002224 eculizumab Drugs 0.000 description 1
- 230000007613 environmental effect Effects 0.000 description 1
- 229940116977 epidermal growth factor Drugs 0.000 description 1
- 229960000403 etanercept Drugs 0.000 description 1
- 230000003203 everyday effect Effects 0.000 description 1
- 229960004222 factor ix Drugs 0.000 description 1
- 229960000301 factor viii Drugs 0.000 description 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- 108700014844 flt3 ligand Proteins 0.000 description 1
- 238000005187 foaming Methods 0.000 description 1
- 239000012737 fresh medium Substances 0.000 description 1
- 230000004927 fusion Effects 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 230000006251 gamma-carboxylation Effects 0.000 description 1
- 229940044627 gamma-interferon Drugs 0.000 description 1
- 229960003297 gemtuzumab ozogamicin Drugs 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 102000018146 globin Human genes 0.000 description 1
- 108060003196 globin Proteins 0.000 description 1
- 229960004666 glucagon Drugs 0.000 description 1
- MASNOZXLGMXCHN-ZLPAWPGGSA-N glucagon Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(O)=O)C(C)C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](C)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CCCCN)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@@H](NC(=O)CNC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CO)NC(=O)[C@@H](N)CC=1NC=NC=1)[C@@H](C)O)[C@@H](C)O)C1=CC=CC=C1 MASNOZXLGMXCHN-ZLPAWPGGSA-N 0.000 description 1
- 125000000291 glutamic acid group Chemical group N[C@@H](CCC(O)=O)C(=O)* 0.000 description 1
- 230000013595 glycosylation Effects 0.000 description 1
- 238000006206 glycosylation reaction Methods 0.000 description 1
- 229960001743 golimumab Drugs 0.000 description 1
- 239000000122 growth hormone Substances 0.000 description 1
- 238000012787 harvest procedure Methods 0.000 description 1
- 108010037896 heparin-binding hemagglutinin Proteins 0.000 description 1
- 230000006801 homologous recombination Effects 0.000 description 1
- 238000002744 homologous recombination Methods 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 102000051284 human FGL2 Human genes 0.000 description 1
- 230000033444 hydroxylation Effects 0.000 description 1
- 238000005805 hydroxylation reaction Methods 0.000 description 1
- 229960001001 ibritumomab tiuxetan Drugs 0.000 description 1
- 229940072221 immunoglobulins Drugs 0.000 description 1
- 238000012606 in vitro cell culture Methods 0.000 description 1
- 229960000598 infliximab Drugs 0.000 description 1
- 229940125396 insulin Drugs 0.000 description 1
- 108010043603 integrin alpha4beta7 Proteins 0.000 description 1
- 108010044426 integrins Proteins 0.000 description 1
- 102000006495 integrins Human genes 0.000 description 1
- 229960003130 interferon gamma Drugs 0.000 description 1
- 229940047122 interleukins Drugs 0.000 description 1
- 201000010982 kidney cancer Diseases 0.000 description 1
- 229950000518 labetuzumab Drugs 0.000 description 1
- 150000002605 large molecules Chemical class 0.000 description 1
- 238000011031 large-scale manufacturing process Methods 0.000 description 1
- 230000002045 lasting effect Effects 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 201000005202 lung cancer Diseases 0.000 description 1
- 208000020816 lung neoplasm Diseases 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 230000014759 maintenance of location Effects 0.000 description 1
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 description 1
- 229950001869 mapatumumab Drugs 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 229950008001 matuzumab Drugs 0.000 description 1
- 238000011177 media preparation Methods 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- 229960005108 mepolizumab Drugs 0.000 description 1
- MYWUZJCMWCOHBA-VIFPVBQESA-N methamphetamine Chemical compound CN[C@@H](C)CC1=CC=CC=C1 MYWUZJCMWCOHBA-VIFPVBQESA-N 0.000 description 1
- 239000007758 minimum essential medium Substances 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 229960001521 motavizumab Drugs 0.000 description 1
- 229960003816 muromonab-cd3 Drugs 0.000 description 1
- 201000000050 myeloid neoplasm Diseases 0.000 description 1
- 229960005027 natalizumab Drugs 0.000 description 1
- 230000001338 necrotic effect Effects 0.000 description 1
- 229940053128 nerve growth factor Drugs 0.000 description 1
- 229940032018 neurotrophin 3 Drugs 0.000 description 1
- 229950010203 nimotuzumab Drugs 0.000 description 1
- 238000010899 nucleation Methods 0.000 description 1
- 235000021232 nutrient availability Nutrition 0.000 description 1
- XXUPLYBCNPLTIW-UHFFFAOYSA-N octadec-7-ynoic acid Chemical compound CCCCCCCCCCC#CCCCCCC(O)=O XXUPLYBCNPLTIW-UHFFFAOYSA-N 0.000 description 1
- 229960002450 ofatumumab Drugs 0.000 description 1
- 229960000470 omalizumab Drugs 0.000 description 1
- 229950007283 oregovomab Drugs 0.000 description 1
- 229960000402 palivizumab Drugs 0.000 description 1
- 201000002528 pancreatic cancer Diseases 0.000 description 1
- 208000008443 pancreatic carcinoma Diseases 0.000 description 1
- 229960001972 panitumumab Drugs 0.000 description 1
- 229960005570 pemtumomab Drugs 0.000 description 1
- 229960002087 pertuzumab Drugs 0.000 description 1
- 239000008194 pharmaceutical composition Substances 0.000 description 1
- 230000035790 physiological processes and functions Effects 0.000 description 1
- 102000040430 polynucleotide Human genes 0.000 description 1
- 108091033319 polynucleotide Proteins 0.000 description 1
- 239000002157 polynucleotide Substances 0.000 description 1
- GCYXWQUSHADNBF-AAEALURTSA-N preproglucagon 78-108 Chemical compound C([C@@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCCCN)C(=O)NCC(=O)N[C@@H](CCCNC(N)=N)C(=O)NCC(O)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCCN)NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CCC(N)=O)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@@H](NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C)NC(=O)[C@@H](N)CC=1N=CNC=1)[C@@H](C)O)[C@@H](C)O)C(C)C)C1=CC=CC=C1 GCYXWQUSHADNBF-AAEALURTSA-N 0.000 description 1
- 238000004886 process control Methods 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 210000002307 prostate Anatomy 0.000 description 1
- 108060006633 protein kinase Proteins 0.000 description 1
- 238000005086 pumping Methods 0.000 description 1
- 229960003876 ranibizumab Drugs 0.000 description 1
- 239000002464 receptor antagonist Substances 0.000 description 1
- 229940044551 receptor antagonist Drugs 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000000241 respiratory effect Effects 0.000 description 1
- 229960004641 rituximab Drugs 0.000 description 1
- 238000005096 rolling process Methods 0.000 description 1
- 229950009092 rovelizumab Drugs 0.000 description 1
- 238000011218 seed culture Methods 0.000 description 1
- 239000012679 serum free medium Substances 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 239000011343 solid material Substances 0.000 description 1
- 230000009870 specific binding Effects 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 230000019635 sulfation Effects 0.000 description 1
- 238000005670 sulfation reaction Methods 0.000 description 1
- 230000003319 supportive effect Effects 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 238000004114 suspension culture Methods 0.000 description 1
- 101150047061 tag-72 gene Proteins 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 210000001519 tissue Anatomy 0.000 description 1
- 239000003104 tissue culture media Substances 0.000 description 1
- 229960000187 tissue plasminogen activator Drugs 0.000 description 1
- 229960003989 tocilizumab Drugs 0.000 description 1
- 229960005267 tositumomab Drugs 0.000 description 1
- 239000011573 trace mineral Substances 0.000 description 1
- 235000013619 trace mineral Nutrition 0.000 description 1
- 230000002463 transducing effect Effects 0.000 description 1
- 230000001131 transforming effect Effects 0.000 description 1
- 229960000575 trastuzumab Drugs 0.000 description 1
- 238000003211 trypan blue cell staining Methods 0.000 description 1
- VBEQCZHXXJYVRD-GACYYNSASA-N uroanthelone Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CS)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CS)C(=O)N[C@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)NCC(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](CO)C(=O)NCC(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CS)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O)C(C)C)[C@@H](C)O)NC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@@H](NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CCSC)NC(=O)[C@H](CS)NC(=O)[C@@H](NC(=O)CNC(=O)CNC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CS)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CS)NC(=O)CNC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@@H](N)CC(N)=O)C(C)C)[C@@H](C)CC)C1=CC=C(O)C=C1 VBEQCZHXXJYVRD-GACYYNSASA-N 0.000 description 1
- 229960003824 ustekinumab Drugs 0.000 description 1
- 239000013598 vector Substances 0.000 description 1
- 231100000747 viability assay Toxicity 0.000 description 1
- 238000003026 viability measurement method Methods 0.000 description 1
- 239000002699 waste material Substances 0.000 description 1
- 229950008250 zalutumumab Drugs 0.000 description 1
- 229950009002 zanolimumab Drugs 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K1/00—General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
- C07K1/14—Extraction; Separation; Purification
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/0018—Culture media for cell or tissue culture
- C12N5/0037—Serum-free medium, which may still contain naturally-sourced components
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0603—Embryonic cells ; Embryoid bodies
- C12N5/0604—Whole embryos; Culture medium therefor
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12P—FERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
- C12P21/00—Preparation of peptides or proteins
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12P—FERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
- C12P21/00—Preparation of peptides or proteins
- C12P21/02—Preparation of peptides or proteins having a known sequence of two or more amino acids, e.g. glutathione
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/10—Immunoglobulins specific features characterized by their source of isolation or production
- C07K2317/14—Specific host cells or culture conditions, e.g. components, pH or temperature
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2500/00—Specific components of cell culture medium
- C12N2500/02—Atmosphere, e.g. low oxygen conditions
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2500/00—Specific components of cell culture medium
- C12N2500/30—Organic components
- C12N2500/32—Amino acids
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2500/00—Specific components of cell culture medium
- C12N2500/99—Serum-free medium
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2510/00—Genetically modified cells
- C12N2510/02—Cells for production
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2523/00—Culture process characterised by temperature
Definitions
- the invention provides a method for culturing mammalian cells.
- the method provides greater control over cell growth to achieve high product titer cell cultures.
- New cell culture methods that provide even incremental improvements in recombinant protein production are valuable, given the expense of large scale cell culture processes and the growing demand for greater quantities of and lower costs for biological products.
- the invention fulfills these needs by providing a simple, easy and inexpensive method of controlling cell growth while increasing protein production.
- the present invention provides a method of arresting cell growth in a mammalian cell culture expressing a recombinant protein comprising establishing a mammalian cell culture in a serum-free culture medium in a bioreactor; inducing cell growth-arrest by perfusion with a serum-free perfusion medium having an L-asparagine concentration of 5mM or less; maintaining the mammalian cells in a growth-arrested state by perfusion with a serum-free perfusion medium having an L-asparagine concentration of 5mM or less.
- the present invention also provides a method of increasing recombinant protein production in a mammalian cell culture expressing a recombinant protein comprising establishing a mammalian cell culture in a serum-free culture medium in a bioreactor; inducing cell growth-arrest by perfusion with a serum-free perfusion medium having an L- asparagine concentration of 5mM or less; maintaining the mammalian cells in a growth- arrested state by perfusion with a serum-free perfusion medium having an L-asparagine concentration of 5mM or less.
- recombinant protein production in the mammalian cell culture is increased compared to a culture where the cells are not subjected to L-asparagine-induced cell growth-arrest.
- the present invention also provides a method of limiting a mammalian cell culture expressing a recombinant protein at a desired packed cell volume comprising establishing a mammalian cell culture in a serum- free culture medium in a bioreactor; inducing cell growth- arrest by perfusion with a serum-free perfusion medium having an L-asparagine concentration of 5mM or less; maintaining the mammalian cells in a growth-arrested state by perfusion with a serum-free perfusion medium having an L-asparagine concentration of 5mM or less.
- any of the methods above begins on or before day 3 of the culture.
- in any of the methods above induction of cell growth-arrest takes place prior to a production phase.
- in any of the methods above induction of cell growth-arrest takes place during a production phase.
- in any of the methods above cell growth- arrest is induced by L-asparagine starvation.
- any of the methods above further comprise a temperature shift from 36°C to 31°C.
- any of the methods above further comprise a temperature shift from 36°C to 33°C.
- the temperature shift occurs at the transition between a growth phase and a production phase. In yet another embodiment the temperature shift occurs during a production phase. In another embodiment the methods above further comprise a packed cell volume during a production phase less than or equal to 35%. In a related embodiment the packed cell volume during a production phase is less than or equal to 35%
- the present invention also provides a method of culturing mammalian cells expressing a recombinant protein comprising; establishing a mammalian cell culture in a serum-free culture medium in a bioreactor; growing the mammalian cells during a growth phase and supplementing the culture medium with bolus feeds of a serum-free feed medium, and maintaining the mammalian cells during a production phase by perfusion with a serum- free perfusion medium, wherein the packed cell volume during the production phase is less than or equal to 35%.
- perfusion begins on or about day 5 to on or about day 9 of the cell culture. In a related embodiment perfusion begins on or about day 5 to on or about day 7 of the cell culture.
- perfusion begins when the cells have reached a production phase.
- the method further comprises inducing cell growth-arrest by L-asparagine starvation followed by perfusion with a serum- free perfusion medium having an L-asparagine concentration of 5 mM or less.
- the method further comprises inducing cell growth-arrest by perfusion with a serum-free perfusion medium having an L-asparagine concentration of 5 mM or less.
- the concentration of L-asparagine in the serum- free perfusion medium is less than or equal to 5mM.
- the concentration of L-asparagine in the serum-free perfusion medium is less than or equal to 4.0 mM.
- the concentration of L-asparagine in the serum-free perfusion medium is less than or equal to 3.0 mM. In still another embodiment the concentration of L- asparagine in the serum-free perfusion medium is less than or equal to 2.0 mM. In yet another embodiment the concentration of L-asparagine in the serum-free perfusion medium is less than or equal to 1.0 mM. In yet another embodiment the concentration of L-asparagine in the serum-free perfusion medium is 0 mM. In another embodiment perfusion is performed at a rate that increases during the production phase from 0.25 working volume per day to 1.0 working volume per day during the cell culture.
- perfusion is performed at a rate that reaches 1.0 working volume per day on day 9 to day 11 of the cell culture. In another related embodiment perfusion is performed at a rate that reaches 1.0 working volume per day on day 10 of the cell culture. In yet another embodiment the bolus feeds of serum- free feed medium begin on day 3 or day 4 of the cell culture.
- the method further comprises a temperature shift from 36°C to 31°C. In another embodiment the method further comprises a temperature shift from 36°C to 33°C. In a related embodiment the temperature shift occurs at the transition between the growth phase and production phase. In a related embodiment the temperature shift occurs during the production phase.
- the L-asparagine concentration of the cell culture medium is monitored prior to and during L-asparagine starvation.
- the packed cell volume is less than or equal to 35%. In a related embodiment the packed cell volume is less than or equal to 30%.
- the viable cell density of the mammalian cell culture at a packed cell volume less than or equal to 35 % is lOxlO 6 viable cells/ml to 80xl0 6 viable cells/ml. In a related embodiment the viable cell density of the mammalian cell culture is 20x10 6 viable cells/ml to 30xl0 6 viable cells/ml.
- perfusion comprises continuous perfusion.
- the rate of perfusion is constant.
- perfusion is performed at a rate of less than or equal to 1.0 working volumes per day.
- the mammalian cell culture is established by inoculating the bioreactor with at least 0.5 x 10 6 to 3.0 x 10 6 cells/mL in a serum-free culture medium.
- the mammalian cell culture is established by inoculating the bioreactor with at least 0.5 x 10 6 to 1.5 x 10 6 cells/mL in a serum- free culture medium.
- the perfusion is accomplished by alternating tangential flow.
- the bioreactor has a capacity of at least 500L. In a related embodiment the bioreactor has a capacity of at least 500L to 2000L. In yet another related embodiment the bioreactor has a capacity of at least 1000L to 2000L.
- the recombinant protein is selected from the group consisting of a human antibody, a humanized antibody, a chimeric antibody, a recombinant fusion protein, or a cytokine.
- any of the methods above further comprise a step of harvesting the recombinant protein produced by the cell culture.
- the recombinant protein produced by the cell culture is purified and formulated in a pharmaceutically acceptable formulation.
- Figure 1A Viable cell density
- Figure IB Viability
- Figure 1C Titer
- Figure 3 1.0 start perfusion volume, no temperature shift: solid circle. 1.0 start perfusion volume, temperature shift: open circle (o). 0.75 start perfusion volume perfusion volume, no temperature shift: solid square ( ⁇ ). 0.75 start perfusion volume, temperature shift: open square ( ⁇ ).
- Figure 4 Batch start with low asparagine amount: open triangle ( ⁇ ). Batch start with control L-asparagine amount: solid triangle (A). Fed-batch start with low L-asparagine amount: open diamond (0). Fed-batch start with control L-asparagine amount: solid diamond ( ⁇ ). Sparge with drilled pipe: Solid line. Sparge with sintered sparger: dashed line.
- Figure 4A Viable Cell Density, Figure 4B Viability, Figure 4C PCV Adjusted Titer
- Figure 5 Cultures grown in medium containing 17.3 mM or 5 mM L-asparagine and 4.6 mM or 10 mM L-glutamine. 17.3mM L-asparagine and 4.6 mM L-glutamine solid diamond ( ⁇ ) or 5 mM L-asparagine, 10 mM L-glutamine open diamond (0).
- Figure 5A Viable cell density.
- Figure 5B Titer.
- Figure 5C Packed cell volume (PCV).
- Figure 5D PCV-adjusted titer.
- Figure 5E Viability.
- Figure 6 Cultures at 2L bench scale and 500L pilot scale, with 5 mM L-asparagine, 10 mM L-glutamine.
- Medium containing 5 mM L-asparagine, 10 mM L-glutamine at 2L bench scale is represented by the solid diamond ( ⁇ ) and the 500L pilot scale is represented by the open diamond (0)
- Figure 6A Viable cell density.
- Figure 6B Titer.
- Figure 6C Packed cell volume (PCV).
- Figure 6D PCV-adjusted titer.
- Figure 6E Viability.
- packed cell volume can be limited to a desired level during the production phase by inducing cell growth-arrest in the cultured cells by exposure to low L-asparagine conditions.
- Cell growth-arrest can be achieved and maintained by using a perfusion culture medium that contains a limiting concentration of L-asparagine and maintaining a low concentration of L- asparagine in the cell culture (5mM or less).
- the growth-arrested cells showed increased productivity when growth-arrest was initiated by low L-asparagine or through L-asparagine starvation and the growth-arrested cells were subsequently maintained with the cell culture and perfusion medium having an L-asparagine concentration of 5mM or less.
- a growth-arrested, high productivity production phase can be achieved by manipulating the concentration of L-asparagine.
- depletion of L- asparagine resulted in growth-arrest.
- the culture was repeatedly starved of L-asparagine despite repeated feedings due to consumption of L-asparagine and/or the conversion to L-aspartate.
- the extracellular L-asparagine can be converted to L-aspartate and ammonia. L-asparagine depletion resulted in cell cycle arrest.
- the feed medium can be switched from a formulation containing a high (growth promoting) level of L- asparagine during bolus feeds to a lower (growth-arresting) level of L-asparagine during perfusion feeding.
- Cell cultures that have been growth-arrested by limiting L-asparagine can be stimulated into a high productivity state by adding back low levels of L-asparagine.
- the method includes inducing cell growth-arrest in a mammalian cell culture by subjecting the cell culture to serum- free medium having an L- asparagine concentration of 5mM or less which includes OmM L-asparageine).
- Such induction can be initiated by L-asparagine starvation or by creating a low L-asparagine environment by perfusing the culture with a serum-free perfusion medium having an L- asparagine concentration of 5mM or less and maintaining the culture in a low L-asparagine environment.
- the cell culture is then maintained in the growth-arrested state by perfusing with a serum-free perfusion medium with an L-asparagine at a concentration of 5mM or less and maintaining the culture in the low L-asparagine environment.
- Mammalian cells maintained in the low asparagine growth-arrested state exhibited greater productivity (g protein/cell/day and g protein/cell mass/day) than those which were not low asparagine growth-arrested.
- Such method is also useful for limiting a mammalian cell culture at a desired packed cell volume.
- Packed cell volume during the production phase could be limited at a desired level by reducing L-asparagine levels in the production culture medium.
- Asparagine concentrations of 5mM or less in the perfusion medium were sufficient to control cell growth during culture and limit to a desired packed cell volume.
- the methods described herein provide greater control over cell growth to high product titer cell cultures; and as such can simplify the gassing strategy compared to a high biomass perfusion processes and minimize product loss during harvest and downstream processing.
- the method begins with establishing a mammalian cell culture in a production bioreactor.
- a production bioreactor Preferably smaller production bioreactors are used, in one embodiment the bioreactors are 500L to 2000L. In a preferred embodiment, 1000L - 2000L bioreactors are used.
- the seed cell density used to inoculate the bioreactor can have a positive impact on the level of recombinant protein produced.
- the bioreactor is inoculated with at least 0.5 xlO 6 up to and beyond 3.0 xlO 6 viable cells/mL in a serum-free culture medium. In a preferred embodiment the inoculation is l .OxlO 6 viable cells/mL.
- the mammalian cells then undergo an exponential growth phase.
- the cell culture can be maintained without supplemental feeding until a desired cell density is achieved.
- the cell culture is maintained for up to 3 days without supplemental feeding followed by perfusion with a serum-free perfusion medium having an L-asparagine concentration of 5mM or less to induce and maintain low L-asparagine growth-arrest.
- the culture can be inoculated at a desired cell density to begin the production phase without a brief growth phase with cell growth-arrest initiated immediately upon inoculation by perfusing the cell culture with serum-free perfusion medium containing 5mM or less L-asparagine to induce and maintain low L-asparagine growth-arrest.
- the switch from the growth phase to production phase can also be initiated by L-asparagine starvation (subjecting cells to a 0 mM L-asparagine environment) followed by perfusion with a cell culture medium having an L-asparagine concentration of equal to or less than 5 mM. and maintaining the concentrating of L-asparagine in the cell culture at that level.
- growth-arrest which may also be referred to as “cell growth- arrest” is the point where cells stop increasing in number or when the cell cycle no longer progresses. Growth-arrest can be monitored by determining the viable cell density of a cell culture. Some cells in a growth-arrested state may increase in size but not number, so the packed cell volume of a growth-arrested culture may increase. Growth-arrest can be reversed to some extent, if the cells are not in declining health, by adding additional L-asparagine to the cell culture.
- L-asparagine starvation occurs when the L-asparagine concentration in a cell culture medium is effectively at OmM. Starvation can result in growth-arrest within 24 hours. Starvation for longer than 48 hours could damage the health of the cells.
- the L-asparagine concentration in the cell culture must be maintained at 5mM or less.
- the cell culture medium concentration of L-asparagine required to arrest cell growth is dependent on the ability of the cells to make their own asparagine. For cultures where cells can make their own asparagine, a lower concentration, or even removal of L-asparagine from the medium may be required for growth-arrest. For cultures that are unable to make their own asparagine, for example, cells that lack active asparagine synthetase enzyme, concentrations above zero up to 5 mM L-asparagine could be used to arrest growth.
- packed cell volume As used herein, "packed cell volume” (PCV), also referred to as “percent packed cell volume” (%PCV), is the ratio of the volume occupied by the cells, to the total volume of cell culture, expressed as a percentage (see Stettler, wt al., (2006) Biotechnol Bioeng. Dec 20:95(6): 1228-33). Packed cell volume is a function of cell density and cell diameter; increases in packed cell volume could arise from increases in either cell density or cell diameter or both. Packed cell volume is a measure of the solid content in the cell culture. Solids are removed during harvest and downstream purification. More solids mean more effort to separate the solid material from the desired product during harvest and downstream purification steps.
- the desired product can become trapped in the solids and lost during the harvest process, resulting in a decreased product yield.
- host cells vary in size and cell cultures also contain dead and dying cells and other cellular debris
- packed cell volume is a more accurate way to describe the solid content within a cell culture than cell density or viable cell density. For example, a 2000L culture having a cell density of 50 x 10 6 cells/ml would have vastly different packed cell volumes depending on the size of the cells.
- some cells when in a growth-arrested state, will increase in size, so the packed cell volume prior to growth-arrest and post growth-arrest will likely be different, due to increase in biomass as a result to cell size increase.
- the percent packed cell volume is equal to or less than 35%.
- the desired packed cell volume maintained during the production phase is equal to or less than 35%.
- the packed cell volume is equal to or less than 30%.
- the packed cell volume is equal to or less than 20%.
- the packed cell volume is equal to or less than 15%.
- the packed cell volume is equal to or less than 10%.
- cell density refers to the number of cells in a given volume of culture medium.
- “Viable cell density” refers to the number of live cells in a given volume of culture medium, as determined by standard viability assays (such as trypan blue dye exclusion method).
- the desired viable cell density at the transition between the growth and production phases and maintained during the production phase is one that provides a packed cell volume of equal to or less than 35%.
- the viable cell density is at least about lOxlO 6 viable cells/mL to 80x10 6 viable cells/mL. In one embodiment the viable cell density is at least about lOxlO 6 viable cells/mL to 70x10 6 viable cells/mL. In one embodiment the viable cell density is at least about lOxlO 6 viable cells/mL to 60x10 6 viable cells/mL. In one embodiment the viable cell density is at least about lOxlO 6 viable cells/mL to 50x10 6 viable cells/mL.
- the viable cell density is at least about lOxlO 6 viable cells/mL to 40x10 6 viable cells/mL. In a preferred embodiment the viable cell density is at least about lOxlO 6 viable cells/mL to 30xl0 6 viable cells/mL. In another preferred embodiment the viable cell density is at least about lOxlO 6 viable cells/mL to 20x10 6 viable cells/mL. In another preferred embodiment, the viable cell density is at least about 20x10 6 viable cells/mL to 30xl0 6 viable cells/mL. In another preferred embodiment the viable cell density is at least about 20x10 6 viable cells/mL to at least about 25x10 6 viable cells/mL, more preferably at least about 20x10 6 viable cells/mL.
- Lower packed cell volume during the production phase helps mitigate dissolved oxygen sparging problems that can hinder higher cell density perfusion cultures.
- the lower packed cell volume also allows for a smaller media volume which allows for the use of smaller media storage vessels and can be combined with slower flow rates.
- Lower packed cell volume also has less impact on harvest and downstream processing, compared to higher cell biomass cultures. All of which reduces the costs associated with manufacturing recombinant protein therapeutics.
- Batch culture a discontinuous method where cells are grown in a fixed volume of culture media for a short period of time followed by a full harvest. Cultures grown using the batch method experience an increase in cell density until a maximum cell density is reached, followed by a decline in viable cell density as the media components are consumed and levels of metabolic by-products (such as lactate and ammonia) accumulate. Harvest typically occurs at the point when the maximum cell density is achieved (typically 5-10xl0 6 cells/mL, depending on media formulation, cell line, etc).
- the batch process is the simplest culture method, however viable cell density is limited by the nutrient availability and once the cells are at maximum density, the culture declines and production decreases. There is no ability to extend a production phase because the accumulation of waste products and nutrient depletion rapidly lead to culture decline, (typically around 3 to 7 days).
- Fed-batch culture improves on the batch process by providing bolus or continuous media feeds to replenish those media components that have been consumed. Since fed-batch cultures receive additional nutrients throughout the run, they have the potential to achieve higher cell densities (>10 to 30xl0 6 cells/ml, depending on media formulation, cell line, etc)) and increased product titers, when compared to the batch method. Unlike the batch process, a biphasic culture can be created and sustained by manipulating feeding strategies and media formulations to distinguish the period of cell proliferation to achieve a desired cell density (the growth phase) from the period of suspended or slow cell growth (the production phase). As such, fed batch cultures have the potential to achieve higher product titers compared to batch cultures.
- a batch method is used during the growth phase and a fed-batch method used during the production phase, but a fed-batch feeding strategy can be used throughout the entire process.
- bioreactor volume is a limiting factor which limits the amount of feed.
- metabolic by-product accumulation will lead to culture decline, which limits the duration of the production phase, about 1.5 to 3 weeks.
- Fed-batch cultures are discontinuous and harvest typically occurs when metabolic by-product levels or culture viability reach predetermined levels.
- Perfusion methods offer potential improvement over the batch and fed-batch methods by adding fresh media and simultaneously removing spent media.
- Typical large scale commercial cell culture strategies strive to reach high cell densities, 60 - 90(+) x 10 6 cells/mL where almost a third to over one-half of the reactor volume is biomass. With perfusion culture, extreme cell densities of >1 x 10 8 cells/mL have been achieved and even higher densities are predicted.
- Typical perfusion cultures begin with a batch culture start-up lasting for a day or two followed by continuous, step-wise and/or intermittent addition of fresh feed media to the culture and simultaneous removal of spend media with the retention of cells and additional high molecular weight compounds such as proteins (based on the filter molecular weight cutoff) throughout the growth and production phases of the culture.
- a large scale cell culture strategy that combines fed batch feeding during the growth phase followed by continuous perfusion during the production phase.
- the method targets a production phase where the cell culture is maintained at a packed cell volume of less than or equal to 35%.
- the method also provides the initiation and maintenance of cell growth-arrest due to low asparagine.
- Fed batch culture is a widely -practiced culture method for large scale production of proteins from mammalian cells. See e.g. Chu and Robinson (2001), Current Opin.
- a fed batch culture of mammalian cells is one in which the culture is fed, either continuously or periodically, with a concentrated feed medium containing nutrients. Feeding can occur on a predetermined schedule of, for example, every day, once every two days, once every three days, etc. When compared to a batch culture, in which no feeding occurs, a fed batch culture can produce greater amounts of recombinant protein. See e.g. U.S. Patent No. 5,672,502.
- a fed-batch culture with bolus feeds is used to maintain a cell culture during the growth phase.
- Perfusion feeding can then be used during a production phase.
- perfusion begins when the cells have reached a production phase.
- perfusion begins on or about day 5 to on or about day 9 of the cell culture.
- perfusion begins on or about day 5 to on or about day 7 of the cell culture.
- the initiation of cell growth-arrest in the fed-batch culture can be initiated by subjecting the fed-batch culture to a period of L-asparagine starvation followed by perfusion with a serum-free perfusion medium having an L-asparagine concentration of 5 mM or less.
- the L-asparagine concentration of the cell culture medium is monitored prior to and during L-asparagine starvation.
- the initiation of cell growth-arrest in the fed-batch culture can be achieved by perfusion with a serum free perfusion medium having an L-asparagine concentration of 5 mM or less.
- Using bolus feeding during the growth phase allows the cells to transition into the production phase, resulting in less dependence on a temperature shift as a means of initiating and controlling the production phase, however a temperature shift of 36°C to 31°C can take place between the growth phase and production phase.
- the shift is from 36°C to 33°C.
- the bioreactor can be inoculated with at least 0.5 xlO 6 up to and beyond 3.0 xlO 6 viable cells/mL in a serum-free culture medium, preferably 1.0x106 viable cells/mL.
- Perfusion culture is one in which the cell culture receives fresh perfusion feed medium while simultaneously removing spent medium.
- Perfusion can be continuous, stepwise, intermittent, or a combination of any or all of any of these. Perfusion rates can be less than a working volume to many working volumes per day.
- the cells are retained in the culture and the spent medium that is removed is substantially free of cells or has significantly fewer cells than the culture. Recombinant proteins expressed by the cell culture can also be retained in the culture.
- Perfusion can be accomplished by a number of means including centrifugation, sedimentation, or filtration, See e.g. Voisard et al., (2003), Biotechnology and Bioengineering 82:751-65.
- a preferred filtration method is alternating tangential flow filtration. Alternating tangential flow is maintained by pumping medium through hollow-fiber filter modules. See e.g. US Patent No. 6,544,424; Furey (2002) Gen. Eng. News. 22 (7), 62-63.
- perfusion flow rate is the amount of media that is passed through (added and removed) from a bioreactor, typically expressed as some portion or multiple of the working volume, in a given time.
- Working volume refers to the amount of bioreactor volume used for cell culture.
- Perfusion flow rate is one working volume or less per day.
- Perfusion feed medium can be formulated to maximize perfusion nutrient concentration to minimize perfusion rate.
- cell culture or “culture” is meant the growth and propagation of cells outside of a multicellular organism or tissue. Suitable culture conditions for mammalian cells are known in the art. See e.g. Animal cell culture: A Practical Approach, D. Rickwood, ed., Oxford University Press, New York (1992). Mammalian cells may be cultured in suspension or while attached to a solid substrate. Fluidized bed bioreactors, hollow fiber bioreactors, roller bottles, shake flasks, or stirred tank bioreactors, with or without microcarriers, can be used. In one embodiment 500L to 2000L bioreactors are used. In a preferred embodiment, 1000L to 2000L bioreactors are used.
- cell culture medium is a media suitable for growth of animal cells, such as mammalian cells, in in vitro cell culture.
- Cell culture media formulations are well known in the art.
- cell culture media are comprised of buffers, salts, carbohydrates, amino acids, vitamins and trace essential elements. "Serum-free” applies to a cell culture medium that does not contain animal sera, such as fetal bovine serum.
- tissue culture media including defined culture media
- any one or a combination of the following cell culture media can be used: RPMI- 1640 Medium, RPMI-1641 Medium, Dulbecco's Modified Eagle's Medium (DMEM), Minimum Essential Medium Eagle, F- 12K Medium, Ham's F12 Medium, Iscove's Modified Dulbecco's Medium, McCoy's 5A Medium, Leibovitz's L-15 Medium, and serum- free media such as EX-CELLTM 300 Series (JRH Biosciences, Lenexa, Kansas), among others. Serum-free versions of such culture media are also available.
- DMEM Dulbecco's Modified Eagle's Medium
- F- 12K Medium Minimum Essential Medium Eagle
- Ham's F12 Medium Ham's F12 Medium
- Iscove's Modified Dulbecco's Medium McCoy's 5A Medium
- Leibovitz's L-15 Medium and serum- free media
- serum-free media such as EX
- Cell culture media may be supplemented with additional or increased concentrations of components such as amino acids, salts, sugars, vitamins, hormones, growth factors, buffers, antibiotics, lipids, trace elements and the like, depending on the requirements of the cells to be cultured and/or the desired cell culture parameters.
- components such as amino acids, salts, sugars, vitamins, hormones, growth factors, buffers, antibiotics, lipids, trace elements and the like, depending on the requirements of the cells to be cultured and/or the desired cell culture parameters.
- Concentrated feed medium may be based on just about any cell culture media formulation. Such a concentrated feed medium can contain most of the components of the cell culture medium at, for example, about 5X, 6X, 7X, 8X, 9X, 10X, 12X, 14X, 16X, 20X, 30X, 50X, lOOx, 200X, 400X, 600X, 800X, or even about 1000X of their normal amount. Concentrated feed media are often used in fed batch culture processes.
- the method according to the present invention may be used to improve the production of recombinant proteins in multiple phase culture processes.
- cells are cultured in two or more distinct phases.
- cells may be cultured first in one or more growth phases, under environmental conditions that maximize cell proliferation and viability, then transferred to a production phase, under conditions that maximize protein production.
- a production phase under conditions that maximize protein production.
- the growth and production phases may be preceded by, or separated by, one or more transition phases.
- the method according to the present invention can be employed at least during the growth and production phase of the final production phase of a commercial cell culture, although it may also be employed in a preceding growth phase.
- a production phase can be conducted at large scale.
- a large scale process can be conducted in a volume of at least about 100, 500, 1000, 2000, 3000, 5000, 7000, 8000, 10,000, 15,000, 20,000 liters.
- production is conducted in 500L, 1000L and/or 2000L bioreactors.
- a growth phase may occur at a higher temperature than a production phase.
- a growth phase may occur at a first temperature from about 35°C to about 38°C
- a production phase may occur at a second temperature from about 29°C to about 37°C, optionally from about 30°C to about 36°C or from about 30°C to about 34°C.
- chemical inducers of protein production such as, for example, caffeine, butyrate, and hexamethylene bisacetamide (HMBA) may be added at the same time as, before, and/or after a temperature shift. If inducers are added after a temperature shift, they can be added from one hour to five days after the temperature shift, optionally from one to two days after the temperature shift. The cell cultures can be maintained for days or even weeks while the cells produce the desired protein(s).
- Samples from the cell culture can be monitored and evaluated using any of the analytical techniques known in the art. Variety of parameters including recombinant protein and medium quality and characteristics can be monitored for the duration of the culture. Samples can be taken and monitored intermittently at a desirable frequency, including continuous monitoring, real time or near real time. In one embodiment the L-asparagine concentration of the cell culture medium is monitored prior to and during L-asparagine starvation.
- the cell cultures that precede the final production culture are used to generate the seed cells that will be used to inoculate the production bioreactor, the N- 1 culture.
- the seed cell density can have a positive impact on the level of recombinant protein produced. Product levels tend to increase with increasing seed density. Improvement in titer is tied not only to higher seed density, but is likely to be influenced by the metabolic and cell cycle state of the cells that are placed into production.
- Seed cells can be produced by any culture method.
- a preferred method is a perfusion culture using alternating tangential flow filtration.
- An N- 1 bioreactor can be run using alternating tangential flow filtration to provide cells at high density to inoculate a production bioreactor.
- the N- 1 stage may be used to grow cells to densities of >90 x 10 6 cells/mL.
- the N-1 bioreactor can be used to generate bolus seed cultures or can be used as a rolling seed stock culture that could be maintained to seed multiple production bioreactors at high seed cell density.
- the duration of the growth stage of production can range from 7 to 14 days and can be designed so as to maintain cells in exponential growth prior to inoculation of the production bioreactor.
- Perfusion rates, medium formulation and timing are optimized to grow cells and deliver them to the production bioreactor in a state that is most conducive to optimizing their production. Seed cell densities of >15 x 10 6 cells/mL can be achieved for seeding production bioreactors. Higher seed cell densities at inoculation can decrease or even eliminate the time needed to reach a desired production density.
- the invention finds particular utility in improving cell growth, viability and/or protein production via cell culture processes.
- the cell lines also referred to as "host cells" used in the invention are genetically engineered to express a polypeptide of commercial or scientific interest.
- Cell lines are typically derived from a lineage arising from a primary culture that can be maintained in culture for an unlimited time. Genetically engineering the cell line involves transfecting, transforming or transducing the cells with a recombinant polynucleotide molecule, and/or otherwise altering (e.g., by homologous recombination and gene activation or fusion of a recombinant cell with a non-recombinant cell) so as to cause the host cell to express a desired recombinant polypeptide.
- Animal cell lines are derived from cells whose progenitors were derived from a multicellular animal.
- One type of animal cell line is a mammalian cell line.
- a wide variety of mammalian cell lines suitable for growth in culture are available from the American Type Culture Collection (Manassas, Va.) and commercial vendors. Examples of cell lines commonly used in the industry include VERO, BHK, HeLa, CV1 (including Cos), MDCK, 293, 3T3, myeloma cell lines (e.g., NSO, NS 1), PC12, WI38 cells, and Chinese hamster ovary (CHO) cells. CHO cells are widely used for the production of complex recombinant proteins, e.g.
- cytokines, clotting factors, and antibodies Brasel et al. (1996), Blood 88:2004-2012; Kaufman et al. (1988), J. Biol Chem 263:6352-6362; McKinnon et al. (1991), J Mol Endocrinol 6:231-239; Wood et al. (1990), J. Immunol. 145:3011-3016).
- DHFR dihydrofolate reductase
- the methods of the invention can be used to culture cells that express recombinant proteins of interest.
- the expressed recombinant proteins may be secreted into the culture medium from which they can be recovered and/or collected.
- the proteins can be purified, or partially purified, from such culture or component (e.g., from culture medium) using known processes and products available from commercial vendors.
- the purified proteins can then be "formulated", meaning buffer exchanged, sterilized, bulk-packaged, and/or packaged for a final user. Suitable formulations for pharmaceutical compositions include those described in Remington's Pharmaceutical Sciences, 18th ed. 1995, Mack Publishing Company, Easton, PA.
- peptide As used herein "peptide,” “polypeptide” and “protein” are used interchangeably throughout and refer to a molecule comprising two or more amino acid residues joined to each other by peptide bonds. Peptides, polypeptides and proteins are also inclusive of modifications including, but not limited to, glycosylation, lipid attachment, sulfation, gamma- carboxylation of glutamic acid residues, hydroxylation and ADP-ribosylation. Polypeptides can be of scientific or commercial interest, including protein-based drugs. Polypeptides include, among other things, antibodies, fusion proteins, and cytokines.
- Peptides, polypeptides and proteins are produced by recombinant animal cell lines using cell culture methods and may be referred to as "recombinant peptide", “recombinant polypeptide” and “recombinant protein”.
- the expressed protein(s) may be produced intracellularly or secreted into the culture medium from which it can be recovered and/or collected.
- TNF tumor necrosis factor
- flt3 ligand WO 94/28391
- erythropoeitin erythropoeitin
- thrombopoeitin thrombopoeitin
- calcitonin IL-2
- angiopoietin-2 angiopoietin-2
- ligand for receptor activator of NF-kappa B (RANKL, WO 01/36637), tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL, WO 97/01633), thymic stroma-derived lymphopoietin, granulocyte colony stimulating factor, granulocyte-macrophage colony stimulating factor (GM-CSF, Australian Patent No.
- mast cell growth factor keratinocyte growth factor
- stem cell growth factor keratinocyte growth factor
- RANTES human fibrinogen- like 2 protein
- growth hormone insulin, insulinotropin, insulin- like growth factors, parathyroid hormone, interferons including a-interferons, ⁇ -interferon, and consensus interferons (US Patent Nos.
- TNFR tumor necrosis factor receptor
- IL-1 receptor antagonists include: both forms of tumor necrosis factor receptor (TNFR, referred to as p55 and p75, US Patent No. 5,395,760 and US Patent No. 5,610,279), Interleukin- 1 (IL- 1) receptors (types I and II; EP Patent No. 0460846, US Patent No. 4,968,607, and US Patent No. 5,767,064,), IL-1 receptor antagonists (US Patent No.
- IL- 1 antagonists or inhibitors US Patent Nos. 5,981,713, 6,096,728, and 5,075,222
- IL-2 receptors IL-4 receptors
- EP Patent No. 0 367 566 and US Patent No. 5,856,296 IL- 15 receptors
- IL- 17 receptors IL-18 receptors
- Fc receptors granulocyte-macrophage colony stimulating factor receptor
- granulocyte colony stimulating factor receptor receptors for oncostatin-M and leukemia inhibitory factor
- receptor activator of NF-kappa B RANK, WO 01/36637 and US Patent No. 6,271,349
- osteoprotegerin US. Patent No. 6,015,938
- receptors for TRAIL including TRAIL receptors 1, 2, 3, and 4
- receptors that comprise death domains such as Fas or Apoptosis-Inducing Receptor (AIR).
- CD proteins proteins comprising all or part of the amino acid sequences of differentiation antigens (referred to as CD proteins) or their ligands or proteins substantially similar to either of these.
- CD proteins are disclosed in Leukocyte Typing VI (Proceedings of the Vlth International Workshop and Conference, Kishimoto, Kikutani et al., eds., Kobe, Japan, 1996). Similar CD proteins are disclosed in subsequent workshops. Examples of such antigens include CD22, CD27, CD30, CD39, CD40, and ligands thereto (CD27 ligand, CD30 ligand, etc.).
- CD antigens are members of the TNF receptor family, which also includes 4 IBB and OX40.
- the ligands are often members of the TNF family, as are 4 IBB ligand and OX40 ligand.
- Enzymatically active proteins or their ligands can also be produced using the invention.
- proteins comprising all or part of one of the following proteins or their ligands or a protein substantially similar to one of these: a disintegrin and metalloproteinase domain family members including TNF-alpha Converting Enzyme, various kinases, glucocerebrosidase, superoxide dismutase, tissue plasminogen activator, Factor VIII, Factor IX, apolipoprotein E, apolipoprotein A-I, globins, an IL-2 antagonist, alpha- 1 antitrypsin, ligands for any of the above-mentioned enzymes, and numerous other enzymes and their ligands.
- a disintegrin and metalloproteinase domain family members including TNF-alpha Converting Enzyme, various kinases, glucocerebrosidase, superoxide dismutase, tissue plasminogen activ
- antibody includes reference to both glycosylated and non-glycosylated immunoglobulins of any isotype or subclass or to an antigen-binding region thereof that competes with the intact antibody for specific binding, unless otherwise specified, including human, humanized, chimeric, multi-specific, monoclonal, polyclonal, and oligomers or antigen binding fragments thereof.
- proteins having an antigen binding fragment or region such as Fab, Fab', F(ab')2, Fv, diabodies, Fd, dAb, maxibodies, single chain antibody molecules, complementarity determining region (CDR) fragments, scFv, diabodies, triabodies, tetrabodies and polypeptides that contain at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding to a target polypeptide.
- antibody is inclusive of, but not limited to, those that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from a host cell transfected to express the antibody.
- antibodies include, but are not limited to, those that recognize any one or a combination of proteins including, but not limited to, the above-mentioned proteins and/or the following antigens: CD2, CD3, CD4, CD8, CDl la, CD14, CD18, CD20, CD22, CD23, CD25, CD33, CD40, CD44, CD52, CD80 (B7.1), CD86 (B7.2), CD 147, IL-la, IL- ⁇ , IL-2, IL-3, IL-7, IL-4, IL-5, IL-8, IL-10, IL-2 receptor, IL-4 receptor, IL-6 receptor, IL-13 receptor, IL- 18 receptor subunits, FGL2, PDGF- ⁇ and analogs thereof (see US Patent Nos.
- VEGF vascular endothelial growth factor
- TGF TGF- 2
- TGF- ⁇ TGF- ⁇
- EGF receptor see US Patent No. 6,235,883
- VEGF receptor hepatocyte growth factor
- osteoprotegerin ligand interferon gamma
- B lymphocyte stimulator BlyS, also known as BAFF, THANK, TALL- 1, and zTNF4; see Do and Chen-Kiang (2002), Cytokine Growth Factor Rev.
- the invention can also be used to produce recombinant fusion proteins comprising, for example, any of the above-mentioned proteins.
- recombinant fusion proteins comprising one of the above-mentioned proteins plus a multimerization domain, such as a leucine zipper, a coiled coil, an Fc portion of an immunoglobulin, or a substantially similar protein, can be produced using the methods of the invention. See e.g. WO94/10308; Lovejoy et al. (1993), Science 259: 1288-1293; Harbury et al. (1993), Science 262: 1401-05; Harbury et al.
- fusion proteins proteins in which a portion of a receptor is fused to an Fc portion of an antibody such as etanercept (a p75 TNFR:Fc), and belatacept (CTLA4:Fc).
- CHO cells expressing a recombinant antibody were inoculated into 2L production bioreactors at lxlO 6 viable cells/mL in a working volume of 1500 ml of a serum- free defined medium for the fed-batch start and 1800mL for the batch start. Cultures were maintained at 36°C, dissolved oxygen (DO) at 30%, agitation at 215 RPM. Glucose was maintained above 0 g/L and below 8 g/L.
- Perfusion (0.75 Vol/day) for the fed-batch cultures. Perfusion was accomplished using an alternating tangential flow perfusion and filtration system (Refine Technologies, Hanover, NJ, 50 kDa hollow fiber filter). Prior to starting perfusion the fed-batch cultures received bolus feeds of a concentrated serum-free defined feed media on day 4 (7.5% of initial working volume) and day 6 (10% initial working volume). Perfusion rates are provided in Table 1.
- Viable cell density (VCD) and viability were determined using Vi-Cell (Beckman Coulter, Brea, CA). Titer was measured by HPLC analysis. Packed cell volume was determined using VoluPAC (Sartorius, Goettingen, Germany).
- a temperature shift (36.0°C to 33.0°C) was applied when the viable cell density exceeded 20 x 10 6 viable cells/mL which was day 7 and day 11 for the batch and fed-batch start conditions respectively.
- the viable cell density for the fed batch was between 27.7 and 30.7 x 10 6 viable cells/mL while VCD of the batch culture was between 22.5 and 27.4 x 10 6 viable cells/mL (Figure 1A).
- Viability of the fed batch culture was between 73.9 and 77.5% while the batch culture viability was between 72.5 and 83.1% ( Figure IB).
- the titer of the fed batch culture was between 15.3 and 16.1 g/L while the batch culture titer was between 10.6 and 12.3 g/L ( Figure 1C).
- variable cell density (IVCD) values were similar for all four cultures by day 15 (approximately 230 x 10 6 cell days/mL), the specific productivity was higher in the fed-batch start-up conditions.
- the fed batch cultures were continued to day 24.
- a titer of 20 g/L was achieved at 20 days.
- CHO cells expressing a recombinant antibody were inoculated into 2L production bioreactors at lxlO 6 viable cells/mL in a working volume of 1500 ml of a serum- free defined medium for the batch start and 1300mL for the fed-batch start. Cultures were maintained at 36°C, DO at 30%, agitation at 215 RPM for batch cultures. The fed-batch culture was agitated at 430RPM. Fed-batch culture was fed to 7 g/L glucose daily prior to perfusion and all cultures were maintained at or above 4 g/L glucose during perfusion.
- Perfusion (alternating tangential flow) was started on day 4 (0.25 Vol/day) for the batch cultures and on day 8 (0.75 Vol/day) for the fed-batch culture. Prior to starting perfusion the fed-batch culture received bolus feeds of a concentrated serum-free defined feed media on day 4 (7.5% of initial working volume) and day 6 (10% initial working volume). Perfusion flow rates settings are provided in Table 2. The cultures were maintained for 21 days.
- a temperature shift (36.0°C to 33.0°C) was applied to the batch cultures on day 6 when the viable cell density exceeded 20 x 10 6 viable cells/mL, as in Example 1.
- the fed batch culture was maintained at 36.0°C for the duration of the culture.
- the batch start-up method cultures had results similar to those described above with the cells reaching approximately 20 to 25 x 10 6 viable cells/mL with no growth after day 10.
- the fed-batch culture reached almost 30 x 10 6 viable cells/mL on day 20 after spending most of the culture duration below 20 x 10 6 viable cells/mL, see Figure 2A.
- the viabilities all remained above 80% until day 10 and then dropping to about 40% by day 20 for the batch start cultures and 60% for the fed-batch culture, see Figure 2B.
- the titer peaked at almost 15 g/L for the batch start cultures, but it reached over 20 g/L for the high agitation fed-batch culture, see Figure 2C.
- the fed-batch start-up with high agitation perfusion culture achieved the highest titer (over 20 g/L) in 20 days, more than 5 g/L higher than the batch start-up cultures, which was similar to the results described above. No negative effects of a higher agitation rate were observed. Maintaining a constant temperature did not appear to negatively impact the fed batch culture.
- CHO cells expressing a recombinant antibody were inoculated into 2L production bioreactors at lxlO 6 cells/mL in a working volume of 1200 ml of a serum- free defined medium. Cultures were maintained at 36°C, DO at 30%. Prior to perfusion, glucose was fed to 7 g/L daily and during perfusion glucose was maintained above 1 g/L. The culture was maintained for 20 days.
- Temperature shift and perfusion rate did not impact viable cell density, see Figure 3A. However, a temperature shift appears to help preserve viability at later time points in a culture. There appears to be a break out between the temperatures shift conditions starting on Day 15 onward. The viability of the temperature shifted cultures dropped more slowly than the cultures that remained at 36°C, see Figure 3B. As for titer, three cultures showed very similar titers on Day 15 (17.1-17.9 g/L) as well as on Day 20 (22-24 g/L), but one culture had a higher titer on Day 15 (21.58 g/L) as well as on Day 20 (28.33 g/L) (see Figure 3C). Neither the temperature nor perfusion rates appeared to have any impact on titer production, suggesting that cultures can be maintained with different perfusion rates.
- This experiment was designed to investigate the effects of perfusion medium asparagine concentrations and perfusion start conditions with either L-asparagine limited or non-limited culture environments on viable cell density during the production phase.
- CHO cells expressing a recombinant antibody were inoculated into 2L production bioreactors at lxl 0 6 cells/mL in a working volume of 1500 ml for both the batch and fed-batch start methods. Cultures were maintained at 36°C, dissolved oxygen (DO) at 30%, agitation at 400 RPM. Sparging was done using either a drilled pipe or a sintered sparger. Glucose was maintained above 0 g/L and below 8 g/L.
- Perfusion (alternating tangential flow) was started on day 3 (0.29 Vol/day) for the batch start "non-asparagine-limited cultures" and on day 7 (0.48 Vol/day) for the fed-batch "asparagine-limited cultures".
- the batch culture medium contained lOmM L-asparagine.
- Prior to starting perfusion the fed-batch cultures received bolus feeds of a concentrated serum-free defined feed media on days 3 and 6 (7% initial working volume) containing 1 13.4mM L-asparagine.
- Perfusion medium asparagine concentrations were either at a control concentration (17.3mM Asn in a serum free defined perfusion medium) or a low concentration (5mM Asn in a serum free defined perfusion medium). Perfusion was carried out as described above. Perfusion rates are provided in Table 4.
- Viable cell density (VCD) and viability were determined using Vi-Cell (Beckman Coulter, Brea, CA). Titer was measured by HPLC analysis. All cultures were maintained at 36.0°C.
- the packed cell volume adjusted titer of the low asparagine fed-batch culture was 17.0 g/L (adjusted for packed cell volume) while the batch culture titer was between 15.4 g/L ( Figure 4C).
- the controls had titers of 10.2 to 12.9 g/L (batch start) and 14.2 to 15.9 g/L (fed-batch start).
- This experiment compares media conditions during perfusion.
- cells were inoculated into a chemically defined batch medium at a working volume of 1.5L, cultured for 3 days and then perfused for 12 days using a chemically defined perfusion medium containing either 17.3 mM L-asparagine and 4.6 mM L-glutamine or 5 mM L-asparagine and 10 mM L-glutamine.
- Perfusion was accomplished using an alternating tangential flow perfusion and filtration system (Refine Technologies, Hanover, NJ) with a 30 kDa hollow fiber filter (GE Healthcare, Little Chalfont, UK). Perfusion was started on day 3 at a rate of 0.3 culture volumes per day. The rate of perfusion was increased on days 4, 9, and 1 1 as indicated in Table 6 below. Cultures were maintained at 36°C, DO at 30%, pH at 7.0, and agitation at 400 rpm.
- VFD Vi-Cell
- Vi-Cell Beckman Coulter, Brea, CA
- Titer was measured by HPLC analysis.
- Packed cell volume was determined using VoluPAC (Sartorius, Goettingen, Germany).
- This example compares the performance of a clonal, antibody-expressing CHO cell line cultured in an ATF perfusion process using asparagine limitation to control growth at bench and pilot scales.
- the bench-scale model utilized 2L bioreactors and the pilot scale was 500L.
- cells were inoculated into a chemically defined batch medium at a working volume of 1.5L and at pilot scale the working volume was about 378L.
- Cells were cultured for 3 days in the batch medium and then perfused for 12 days using a chemically defined perfusion medium containing 5 mM L-asparagine and 10 mM L-glutamine.
- Perfusion was accomplished using an alternating tangential flow perfusion and filtration system (Refine Technologies, Hanover, NJ) with a 30 kDa hollow fiber filter (GE Healthcare, Little Chalfont, UK). Perfusion was started on day 3 at a rate of 0.3 culture volumes per day. The rate of perfusion was increased on days 4, 9, and 1 1 as indicated in Table 7 below. Cultures were maintained at 36°C, 30% DO, and pH 6.9.
- Viable cell density (VCD) and viability were determined bench scale using Vi-Cell (Beckman Coulter, Brea, CA) and at pilot scale using a CEDEX (Roche Applied Science, Indianapolis, IN). Titer was measured by HPLC analysis. Packed cell volume was determined using VoluPAC (Sartorius, Goettingen, Germany). Table 6. Perfusion rate schedule
Landscapes
- Life Sciences & Earth Sciences (AREA)
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- Wood Science & Technology (AREA)
- Zoology (AREA)
- Biotechnology (AREA)
- Biomedical Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Health & Medical Sciences (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Microbiology (AREA)
- Molecular Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Cell Biology (AREA)
- Medicinal Chemistry (AREA)
- Biophysics (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Immunology (AREA)
- Gynecology & Obstetrics (AREA)
- Developmental Biology & Embryology (AREA)
- Reproductive Health (AREA)
- Analytical Chemistry (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Preparation Of Compounds By Using Micro-Organisms (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Peptides Or Proteins (AREA)
Abstract
Description
Claims
Priority Applications (39)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
ES12738674.6T ES2625045T3 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
IL310968A IL310968A (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
CA2838695A CA2838695C (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
DK12738674.6T DK2726600T3 (en) | 2011-07-01 | 2012-06-29 | Mammalian Cell Culture |
LTEP12738674.6T LT2726600T (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
BR122021007265-5A BR122021007265B1 (en) | 2011-07-01 | 2012-06-29 | Methods for culturing cho mammalian cells expressing a recombinant protein |
KR1020237043604A KR20240005106A (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
KR1020147001985A KR101662412B1 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
JP2014519184A JP5897708B2 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
AU2012279230A AU2012279230B2 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
BR112013033730-3A BR112013033730B1 (en) | 2011-07-01 | 2012-06-29 | Method for culturing mammalian cells expressing recombinant protein |
PL14187232T PL2837680T3 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
IL297998A IL297998B2 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
CN201711406697.2A CN107988166B (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
EP12738674.6A EP2726600B1 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
KR1020207012533A KR102358951B1 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
US14/127,050 US11292829B2 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
KR1020227003428A KR102558247B1 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
EA201391826A EA039023B1 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
KR1020187012639A KR102108663B1 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
EP14187232.5A EP2837680B1 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
CN201280032874.4A CN103827292B (en) | 2011-07-01 | 2012-06-29 | Mammaliancellculture |
SI201230945A SI2726600T1 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
IL288509A IL288509B2 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
MX2013014937A MX351974B (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture. |
KR1020167015700A KR101857380B1 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
KR1020237024499A KR20230114317A (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
IL229803A IL229803B (en) | 2011-07-01 | 2013-12-05 | Mammalian cell culture |
ZA2013/09372A ZA201309372B (en) | 2011-07-01 | 2013-12-10 | Mammalian cell culture |
HK14110449A HK1197081A1 (en) | 2011-07-01 | 2014-10-20 | Mammalian cell culture |
CY20171100366T CY1118863T1 (en) | 2011-07-01 | 2017-03-22 | Mammalian Cultivation |
CY20201100473T CY1123146T1 (en) | 2011-07-01 | 2020-05-25 | MAMMALIAN CELL CULTURE |
US17/018,557 US11634476B2 (en) | 2011-07-01 | 2020-09-11 | Mammalian cell culture |
US17/018,564 US11673941B2 (en) | 2011-07-01 | 2020-09-11 | Mammalian cell culture |
IL278430A IL278430B (en) | 2011-07-01 | 2020-11-02 | Mammalian cell culture |
US17/686,766 US20220185869A1 (en) | 2011-07-01 | 2022-03-04 | Mammalian cell culture |
US18/066,251 US11685772B2 (en) | 2011-07-01 | 2022-12-14 | Mammalian cell culture |
US18/068,922 US20230129523A1 (en) | 2011-07-01 | 2022-12-20 | Mammalian cell culture |
US18/180,828 US11827692B2 (en) | 2011-07-01 | 2023-03-08 | Mammalian cell culture |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201161503737P | 2011-07-01 | 2011-07-01 | |
US61/503,737 | 2011-07-01 |
Related Child Applications (4)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US14/127,050 A-371-Of-International US11292829B2 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
US17/018,557 Division US11634476B2 (en) | 2011-07-01 | 2020-09-11 | Mammalian cell culture |
US17/018,564 Division US11673941B2 (en) | 2011-07-01 | 2020-09-11 | Mammalian cell culture |
US17/686,766 Continuation US20220185869A1 (en) | 2011-07-01 | 2022-03-04 | Mammalian cell culture |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2013006479A2 true WO2013006479A2 (en) | 2013-01-10 |
WO2013006479A3 WO2013006479A3 (en) | 2013-03-28 |
Family
ID=46579326
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2012/045070 WO2013006479A2 (en) | 2011-07-01 | 2012-06-29 | Mammalian cell culture |
Country Status (24)
Country | Link |
---|---|
US (7) | US11292829B2 (en) |
EP (2) | EP2837680B1 (en) |
JP (1) | JP5897708B2 (en) |
KR (7) | KR102558247B1 (en) |
CN (2) | CN103827292B (en) |
AU (5) | AU2012279230B2 (en) |
BR (2) | BR122021007265B1 (en) |
CA (2) | CA2952347A1 (en) |
CL (2) | CL2013003792A1 (en) |
CY (2) | CY1118863T1 (en) |
DK (2) | DK2837680T3 (en) |
EA (1) | EA039023B1 (en) |
ES (2) | ES2788132T3 (en) |
HK (2) | HK1197081A1 (en) |
HU (2) | HUE033279T2 (en) |
IL (5) | IL288509B2 (en) |
LT (2) | LT2837680T (en) |
MX (1) | MX351974B (en) |
PL (2) | PL2726600T3 (en) |
PT (2) | PT2837680T (en) |
SG (1) | SG10201702537RA (en) |
SI (2) | SI2726600T1 (en) |
WO (1) | WO2013006479A2 (en) |
ZA (1) | ZA201309372B (en) |
Cited By (45)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2014109858A1 (en) * | 2013-01-14 | 2014-07-17 | Amgen Inc. | Methods of using cell-cycle inhibitors to modulate one or more properties of a cell culture |
WO2014159259A1 (en) | 2013-03-14 | 2014-10-02 | Amgen Inc. | Methods for increasing mannose content of recombinant proteins |
US8895709B2 (en) | 2008-10-20 | 2014-11-25 | Abbvie Inc. | Isolation and purification of antibodies using protein A affinity chromatography |
US8906646B2 (en) | 2006-09-13 | 2014-12-09 | Abbvie Inc. | Fed-batch method of making human anti-TNF-alpha antibody |
US8911964B2 (en) | 2006-09-13 | 2014-12-16 | Abbvie Inc. | Fed-batch method of making human anti-TNF-alpha antibody |
US8921526B2 (en) | 2013-03-14 | 2014-12-30 | Abbvie, Inc. | Mutated anti-TNFα antibodies and methods of their use |
US8946395B1 (en) | 2013-10-18 | 2015-02-03 | Abbvie Inc. | Purification of proteins using hydrophobic interaction chromatography |
US9017687B1 (en) | 2013-10-18 | 2015-04-28 | Abbvie, Inc. | Low acidic species compositions and methods for producing and using the same using displacement chromatography |
US9062106B2 (en) | 2011-04-27 | 2015-06-23 | Abbvie Inc. | Methods for controlling the galactosylation profile of recombinantly-expressed proteins |
WO2015095809A1 (en) | 2013-12-20 | 2015-06-25 | Biogen Idec Ma Inc. | Use of perfusion seed cultures to improve biopharmaceutical fed-batch production capacity and product quality |
US9067990B2 (en) | 2013-03-14 | 2015-06-30 | Abbvie, Inc. | Protein purification using displacement chromatography |
WO2015105609A1 (en) | 2014-01-13 | 2015-07-16 | Amgen Inc. | Regulating ornithine metabolism to manipulate the high mannose glycoform content of recombinant proteins |
US9085618B2 (en) | 2013-10-18 | 2015-07-21 | Abbvie, Inc. | Low acidic species compositions and methods for producing and using the same |
US9109010B2 (en) | 2008-10-20 | 2015-08-18 | Abbvie Inc. | Viral inactivation during purification of antibodies cross reference to related applications |
US9150645B2 (en) | 2012-04-20 | 2015-10-06 | Abbvie, Inc. | Cell culture methods to reduce acidic species |
US9181337B2 (en) | 2013-10-18 | 2015-11-10 | Abbvie, Inc. | Modulated lysine variant species compositions and methods for producing and using the same |
US9181572B2 (en) | 2012-04-20 | 2015-11-10 | Abbvie, Inc. | Methods to modulate lysine variant distribution |
US9193787B2 (en) | 2012-04-20 | 2015-11-24 | Abbvie Inc. | Human antibodies that bind human TNF-alpha and methods of preparing the same |
US9206390B2 (en) | 2012-09-02 | 2015-12-08 | Abbvie, Inc. | Methods to control protein heterogeneity |
WO2015188009A1 (en) | 2014-06-04 | 2015-12-10 | Amgen Inc. | Methods for harvesting mammalian cell cultures |
US9234033B2 (en) | 2012-09-02 | 2016-01-12 | Abbvie, Inc. | Methods to control protein heterogeneity |
US9249182B2 (en) | 2012-05-24 | 2016-02-02 | Abbvie, Inc. | Purification of antibodies using hydrophobic interaction chromatography |
JP2016514475A (en) * | 2013-03-26 | 2016-05-23 | コヒラス・バイオサイエンシズ・インコーポレイテッド | Protein production method |
WO2016089919A1 (en) | 2014-12-01 | 2016-06-09 | Amgen Inc. | Process for manipulating the level of glycan content of a glycoprotein |
US9481901B2 (en) | 2013-05-30 | 2016-11-01 | Amgen Inc. | Methods for increasing mannose content of recombinant proteins |
JP2016534732A (en) * | 2013-10-31 | 2016-11-10 | アムジエン・インコーポレーテツド | Use of monensin to regulate glycosylation of recombinant proteins |
US9499614B2 (en) | 2013-03-14 | 2016-11-22 | Abbvie Inc. | Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides |
US9550826B2 (en) | 2013-11-15 | 2017-01-24 | Abbvie Inc. | Glycoengineered binding protein compositions |
US9598667B2 (en) | 2013-10-04 | 2017-03-21 | Abbvie Inc. | Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins |
JP2017521057A (en) * | 2014-06-09 | 2017-08-03 | ジェンザイム・コーポレーション | Seed train method and its use |
WO2017149065A1 (en) | 2016-03-02 | 2017-09-08 | Lonza Ltd | Improved fermentation process |
JP2018500041A (en) * | 2014-12-31 | 2018-01-11 | エルジー・ケム・リミテッド | Methods for regulating glycosylation of recombinant glycoproteins |
US10047141B2 (en) | 2014-01-30 | 2018-08-14 | Coherus Biosciences, Inc. | Method of manufacturing a protein by perfusion in media with a low amino acid concentration |
US10213508B2 (en) | 2011-10-18 | 2019-02-26 | Coherus Biosciences, Inc. | Etanercept formulations stabilized with xylitol |
US10493151B2 (en) | 2011-10-18 | 2019-12-03 | Coherus Biosciences, Inc. | Etanercept formulations stabilized with sodium chloride |
US10822429B2 (en) | 2012-07-09 | 2020-11-03 | Coherus Biosciences, Inc. | Etanercept formulations exhibiting marked reduction in sub-visible particles |
US10947306B2 (en) | 2012-09-11 | 2021-03-16 | Coherus Biosciences, Inc. | Correctly folded etanercept in high purity and excellent yield |
US11060058B2 (en) | 2014-06-06 | 2021-07-13 | Genzyme Corporation | Perfusion culturing methods and uses thereof |
WO2022047108A1 (en) * | 2020-08-31 | 2022-03-03 | Regeneron Pharmaceuticals, Inc. | Asparagine feed strategies to improve cell culture performance and mitigate asparagine sequence variants |
WO2022036275A3 (en) * | 2020-08-14 | 2022-05-12 | Bristol-Myers Squibb Company | Manufacturing process for protein |
US11390663B2 (en) | 2013-10-11 | 2022-07-19 | Regeneron Pharmaceuticals, Inc. | Metabolically optimized cell culture |
US11702628B2 (en) | 2017-03-31 | 2023-07-18 | Boehringer Ingelheim International Gmbh | Perfusion medium for culturing mammalian cells and reducing cell bleed in a perfusion cell culture |
WO2024054414A1 (en) | 2022-09-06 | 2024-03-14 | Amgen Inc. | Lean perfusion cell culture methods |
WO2024059235A2 (en) | 2022-09-16 | 2024-03-21 | Amgen Inc. | A method for harvesting products from perfusion cultures |
US12084686B2 (en) | 2018-05-01 | 2024-09-10 | Amgen Inc. | Antibodies with modulated glycan profiles |
Families Citing this family (17)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
KR102558247B1 (en) * | 2011-07-01 | 2023-07-24 | 암젠 인크 | Mammalian cell culture |
JP6530171B2 (en) * | 2014-09-09 | 2019-06-12 | 旭化成メディカル株式会社 | Method of collecting culture product |
CN105385731B (en) * | 2015-12-25 | 2018-10-30 | 上海莱士血液制品股份有限公司 | A kind of perfusion cultural method of eight factors of expression recombination |
WO2017120359A1 (en) * | 2016-01-06 | 2017-07-13 | Oncobiologics, Inc. | Reduction of high molecular weight species, acidic charge species, and fragments in a monoclonal antibody composition |
CN109563161A (en) | 2016-02-03 | 2019-04-02 | 安口生物公司 | For improving the buffer preparation of Antibody stability |
EP3601584A1 (en) * | 2017-03-31 | 2020-02-05 | Boehringer Ingelheim International GmbH | Perfusion medium |
MA49979A (en) | 2017-08-09 | 2020-06-17 | Juno Therapeutics Inc | PROCESSES FOR THE PRODUCTION OF GENETICALLY MODIFIED CELL COMPOSITIONS AND RELATED COMPOSITIONS |
EP3692358A1 (en) | 2017-10-06 | 2020-08-12 | Lonza Ltd | Automated control of cell culture using raman spectroscopy |
CN111212899B (en) | 2017-10-16 | 2024-07-02 | 里珍纳龙药品有限公司 | Perfusion bioreactor and related methods of use |
JP2021505168A (en) * | 2017-12-08 | 2021-02-18 | ジュノー セラピューティクス インコーポレイテッド | Methods for Producing Manipulated T Cell Compositions |
SI3818078T1 (en) * | 2018-07-03 | 2024-05-31 | Bristol-Myers Squibb Company | Methods of producing recombinant proteins |
JP2022516516A (en) * | 2018-12-31 | 2022-02-28 | レプリゲン・コーポレーション | Filters for perfusion and purification of mammalian cell cultures with hydrophobic hollow fibers |
WO2021058713A1 (en) * | 2019-09-27 | 2021-04-01 | Boehringer Ingelheim International Gmbh | Concentrated perfusion medium |
WO2022066595A1 (en) | 2020-09-22 | 2022-03-31 | Bristol-Myers Squibb Company | Methods for producing therapeutic proteins |
CN114574430A (en) * | 2022-03-22 | 2022-06-03 | 成都博宠生物科技有限公司 | Expi293F cell culture method |
CN115287251A (en) * | 2022-08-25 | 2022-11-04 | 无锡药明生物技术股份有限公司 | Intermittent perfusion combined batch feeding culture |
CN116790483B (en) * | 2023-08-22 | 2023-10-20 | 苏州依科赛生物科技股份有限公司 | CHO cell serum-free medium and application thereof |
Citations (24)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4695623A (en) | 1982-05-06 | 1987-09-22 | Amgen | Consensus human leukocyte interferon |
AU588819B2 (en) | 1984-10-29 | 1989-09-28 | Immunex Corporation | Cloning of human granulocyte-macrophage colony stimulating factor gene |
EP0367566A1 (en) | 1988-10-31 | 1990-05-09 | Immunex Corporation | Interleukin-4 receptors |
US4968607A (en) | 1987-11-25 | 1990-11-06 | Immunex Corporation | Interleukin-1 receptors |
EP0460846A1 (en) | 1990-06-05 | 1991-12-11 | Immunex Corporation | Type II interleukin-1 receptors |
US5075222A (en) | 1988-05-27 | 1991-12-24 | Synergen, Inc. | Interleukin-1 inhibitors |
US5149792A (en) | 1989-12-19 | 1992-09-22 | Amgen Inc. | Platelet-derived growth factor B chain analogs |
US5272064A (en) | 1989-12-19 | 1993-12-21 | Amgen Inc. | DNA molecules encoding platelet-derived growth factor B chain analogs and method for expression thereof |
WO1994010308A1 (en) | 1992-10-23 | 1994-05-11 | Immunex Corporation | Methods of preparing soluble, oligomeric proteins |
WO1994028391A1 (en) | 1993-05-24 | 1994-12-08 | Immunex Corporation | Ligands for flt3 receptors |
US5395760A (en) | 1989-09-05 | 1995-03-07 | Immunex Corporation | DNA encoding tumor necrosis factor-α and -β receptors |
WO1997001633A1 (en) | 1995-06-29 | 1997-01-16 | Immunex Corporation | Cytokine that induces apoptosis |
US5610279A (en) | 1989-09-12 | 1997-03-11 | Hoffman-La Roche Inc. | Human TNF receptor |
US5672502A (en) | 1985-06-28 | 1997-09-30 | Celltech Therapeutics Limited | Animal cell culture |
US5767064A (en) | 1990-06-05 | 1998-06-16 | Immunex Corporation | Soluble type II interleukin-1 receptors and methods |
US5981713A (en) | 1994-10-13 | 1999-11-09 | Applied Research Systems Ars Holding N.V. | Antibodies to intereleukin-1 antagonists |
US6015938A (en) | 1995-12-22 | 2000-01-18 | Amgen Inc. | Osteoprotegerin |
US6096728A (en) | 1996-02-09 | 2000-08-01 | Amgen Inc. | Composition and method for treating inflammatory diseases |
US6204363B1 (en) | 1989-10-16 | 2001-03-20 | Amgen Inc. | Stem cell factor |
US6235883B1 (en) | 1997-05-05 | 2001-05-22 | Abgenix, Inc. | Human monoclonal antibodies to epidermal growth factor receptor |
WO2001036637A1 (en) | 1999-11-17 | 2001-05-25 | Immunex Corporation | Receptor activator of nf-kappa b |
US6271349B1 (en) | 1996-12-23 | 2001-08-07 | Immunex Corporation | Receptor activator of NF-κB |
US6337072B1 (en) | 1998-04-03 | 2002-01-08 | Hyseq, Inc. | Interleukin-1 receptor antagonist and recombinant production thereof |
US6544424B1 (en) | 1999-12-03 | 2003-04-08 | Refined Technology Company | Fluid filtration system |
Family Cites Families (40)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4499064A (en) | 1982-06-03 | 1985-02-12 | Clayton Foundation For Research | Assessment of nutritional status of individuals |
US4816401A (en) * | 1985-09-09 | 1989-03-28 | University Of Rochester | Serum free cell culture medium |
US5045468A (en) | 1986-12-12 | 1991-09-03 | Cell Enterprises, Inc. | Protein-free culture medium which promotes hybridoma growth |
US5143842A (en) * | 1988-11-01 | 1992-09-01 | The University Of Colorado Foundation, Inc. | Media for normal human muscle satellite cells |
US5292655A (en) * | 1990-01-29 | 1994-03-08 | Wille Jr John J | Method for the formation of a histologically-complete skin substitute |
US7037721B1 (en) * | 1990-01-29 | 2006-05-02 | Hy-Gene Biomedical, Inc. | Protein-free defined media for the growth of normal human keratinocytes |
GB9118664D0 (en) | 1991-08-30 | 1991-10-16 | Celltech Ltd | Cell culture |
US6310185B1 (en) | 1994-03-08 | 2001-10-30 | Memorial Sloan Kettering Cancer Center | Recombinant human anti-Lewis Y antibodies |
US5856179A (en) | 1994-03-10 | 1999-01-05 | Genentech, Inc. | Polypeptide production in animal cell culture |
JP2000517188A (en) | 1996-08-30 | 2000-12-26 | ライフ テクノロジーズ,インコーポレイテッド | Serum-free mammalian cell culture medium and uses thereof |
US20020012991A1 (en) | 1997-04-07 | 2002-01-31 | Florence Chua Nee Ho Kit Fong | Cell culture media for enhanced protein production |
US20030036505A1 (en) | 2000-09-25 | 2003-02-20 | Human Genome Sciences, Inc. | Signal transduction pathway component polynucleotides, polypeptides, antibodies and methods based thereon |
EP1404813A4 (en) | 2001-06-13 | 2004-11-24 | Genentech Inc | Methods of culturing animal cells and polypeptide production in animal cells |
US7320789B2 (en) | 2001-09-26 | 2008-01-22 | Wyeth | Antibody inhibitors of GDF-8 and uses thereof |
AU2003220529B2 (en) | 2002-03-27 | 2006-09-07 | Immunex Corporation | Methods for increasing polypeptide production |
EP2308958A3 (en) * | 2004-03-05 | 2011-08-10 | DSM IP Assets B.V. | Process for cell culturing by continuous perfusion and alternating tangential flow |
CN1332024C (en) * | 2004-06-09 | 2007-08-15 | 中国科学院大连化学物理研究所 | 3D culture method for micro carrier of sponge cell |
DK2156852T3 (en) | 2004-07-07 | 2011-08-08 | Coloplast As | Catheter comprising Estane 58212 |
US7294484B2 (en) | 2004-08-27 | 2007-11-13 | Wyeth Research Ireland Limited | Production of polypeptides |
TWI374935B (en) * | 2004-08-27 | 2012-10-21 | Pfizer Ireland Pharmaceuticals | Production of α-abeta |
CA2585547A1 (en) * | 2004-10-29 | 2006-05-11 | Centocor, Inc. | Chemically defined media compositions |
US7691611B2 (en) * | 2005-06-03 | 2010-04-06 | Ares Trading S.A. | Production of recombinant IL-18 binding protein |
MX340242B (en) | 2006-07-14 | 2016-07-01 | Dpx Holdings Bv | Improved process for the culturing of cells. |
WO2008024769A2 (en) * | 2006-08-21 | 2008-02-28 | Raven Biotechnologies, Inc. | Intensified perfusion production method |
SI2078071T1 (en) | 2006-11-08 | 2015-05-29 | Wyeth Llc | Rationally designed media for cell culture |
WO2008154014A2 (en) | 2007-06-11 | 2008-12-18 | Amgen Inc. | A method for culturing mammalian cells to improve recombinant protein production |
EP2188371B1 (en) | 2007-08-09 | 2017-12-20 | Wyeth LLC | Use of perfusion to enhance production of fed-batch cell culture in bioreactors |
AU2009290563A1 (en) | 2008-09-15 | 2010-03-18 | Genentech, Inc. | Compositions and methods for regulating cell osmolarity |
CN101603026B (en) * | 2009-06-19 | 2011-01-19 | 华东理工大学 | Animal origin-free low-protein culture medium suitable for animal cell product production |
US8580554B2 (en) | 2009-07-31 | 2013-11-12 | Baxter International Inc. | Method of producing a polypeptide or virus of interest in a continuous cell culture |
KR101574056B1 (en) | 2009-07-31 | 2015-12-02 | 백스터 인터내셔널 인코포레이티드 | Cell culture medium for adamts protein expression |
SG10201901417UA (en) | 2009-08-11 | 2019-03-28 | Genentech Inc | Production of proteins in glutamine-free cell culture media |
HUE035508T2 (en) | 2009-11-17 | 2018-05-02 | Squibb & Sons Llc | Methods for enhanced protein production |
WO2011065940A1 (en) | 2009-11-24 | 2011-06-03 | Biogen Idec Ma Inc. | Method of supplementing culture media to prevent undesirable amino acid substitutions |
MX347981B (en) * | 2010-11-01 | 2017-05-22 | Symphogen As | Pan-her antibody composition. |
WO2012145682A1 (en) * | 2011-04-21 | 2012-10-26 | Amgen Inc. | A method for culturing mammalian cells to improve recombinant protein production |
KR102558247B1 (en) * | 2011-07-01 | 2023-07-24 | 암젠 인크 | Mammalian cell culture |
TWI625390B (en) | 2013-03-14 | 2018-06-01 | 安美基公司 | Methods for increasing mannose content of recombinant proteins |
MX2016005572A (en) | 2013-10-31 | 2016-12-09 | Amgen Inc | Use of monensin to regulate glycosylation of recombinant proteins. |
AU2016357567B2 (en) * | 2015-11-17 | 2020-05-07 | Pfizer Inc. | Media and fermentation methods for producing polysaccharides in bacterial cell culture |
-
2012
- 2012-06-29 KR KR1020227003428A patent/KR102558247B1/en active IP Right Grant
- 2012-06-29 EA EA201391826A patent/EA039023B1/en unknown
- 2012-06-29 EP EP14187232.5A patent/EP2837680B1/en active Active
- 2012-06-29 PL PL12738674T patent/PL2726600T3/en unknown
- 2012-06-29 MX MX2013014937A patent/MX351974B/en active IP Right Grant
- 2012-06-29 US US14/127,050 patent/US11292829B2/en active Active
- 2012-06-29 HU HUE12738674A patent/HUE033279T2/en unknown
- 2012-06-29 CA CA2952347A patent/CA2952347A1/en not_active Abandoned
- 2012-06-29 SI SI201230945A patent/SI2726600T1/en unknown
- 2012-06-29 ES ES14187232T patent/ES2788132T3/en active Active
- 2012-06-29 KR KR1020237043604A patent/KR20240005106A/en not_active Application Discontinuation
- 2012-06-29 JP JP2014519184A patent/JP5897708B2/en active Active
- 2012-06-29 PL PL14187232T patent/PL2837680T3/en unknown
- 2012-06-29 ES ES12738674.6T patent/ES2625045T3/en active Active
- 2012-06-29 BR BR122021007265-5A patent/BR122021007265B1/en active IP Right Grant
- 2012-06-29 CN CN201280032874.4A patent/CN103827292B/en active Active
- 2012-06-29 LT LTEP14187232.5T patent/LT2837680T/en unknown
- 2012-06-29 PT PT141872325T patent/PT2837680T/en unknown
- 2012-06-29 SG SG10201702537RA patent/SG10201702537RA/en unknown
- 2012-06-29 IL IL288509A patent/IL288509B2/en unknown
- 2012-06-29 IL IL297998A patent/IL297998B2/en unknown
- 2012-06-29 LT LTEP12738674.6T patent/LT2726600T/en unknown
- 2012-06-29 PT PT127386746T patent/PT2726600T/en unknown
- 2012-06-29 AU AU2012279230A patent/AU2012279230B2/en active Active
- 2012-06-29 CA CA2838695A patent/CA2838695C/en not_active Expired - Fee Related
- 2012-06-29 CN CN201711406697.2A patent/CN107988166B/en active Active
- 2012-06-29 KR KR1020237024499A patent/KR20230114317A/en active IP Right Grant
- 2012-06-29 KR KR1020167015700A patent/KR101857380B1/en active IP Right Grant
- 2012-06-29 KR KR1020207012533A patent/KR102358951B1/en active IP Right Grant
- 2012-06-29 WO PCT/US2012/045070 patent/WO2013006479A2/en active Application Filing
- 2012-06-29 BR BR112013033730-3A patent/BR112013033730B1/en active IP Right Grant
- 2012-06-29 KR KR1020187012639A patent/KR102108663B1/en active IP Right Grant
- 2012-06-29 DK DK14187232.5T patent/DK2837680T3/en active
- 2012-06-29 IL IL310968A patent/IL310968A/en unknown
- 2012-06-29 KR KR1020147001985A patent/KR101662412B1/en active IP Right Grant
- 2012-06-29 DK DK12738674.6T patent/DK2726600T3/en active
- 2012-06-29 EP EP12738674.6A patent/EP2726600B1/en not_active Revoked
- 2012-06-29 HU HUE14187232A patent/HUE049119T2/en unknown
- 2012-06-29 SI SI201231761T patent/SI2837680T1/en unknown
-
2013
- 2013-12-05 IL IL229803A patent/IL229803B/en active IP Right Grant
- 2013-12-10 ZA ZA2013/09372A patent/ZA201309372B/en unknown
- 2013-12-30 CL CL2013003792A patent/CL2013003792A1/en unknown
-
2014
- 2014-10-20 HK HK14110449A patent/HK1197081A1/en unknown
- 2014-10-20 HK HK15106530.7A patent/HK1206057A1/en unknown
-
2016
- 2016-05-18 CL CL2016001190A patent/CL2016001190A1/en unknown
- 2016-10-12 AU AU2016244255A patent/AU2016244255A1/en not_active Abandoned
-
2017
- 2017-03-22 CY CY20171100366T patent/CY1118863T1/en unknown
-
2018
- 2018-06-05 AU AU2018203972A patent/AU2018203972B2/en active Active
-
2020
- 2020-05-25 CY CY20201100473T patent/CY1123146T1/en unknown
- 2020-09-11 US US17/018,564 patent/US11673941B2/en active Active
- 2020-09-11 US US17/018,557 patent/US11634476B2/en active Active
- 2020-11-02 IL IL278430A patent/IL278430B/en unknown
- 2020-12-03 AU AU2020281075A patent/AU2020281075B2/en active Active
-
2022
- 2022-03-04 US US17/686,766 patent/US20220185869A1/en active Pending
- 2022-12-14 US US18/066,251 patent/US11685772B2/en active Active
- 2022-12-20 US US18/068,922 patent/US20230129523A1/en active Pending
-
2023
- 2023-02-24 AU AU2023201127A patent/AU2023201127A1/en active Pending
- 2023-03-08 US US18/180,828 patent/US11827692B2/en active Active
Patent Citations (26)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4695623A (en) | 1982-05-06 | 1987-09-22 | Amgen | Consensus human leukocyte interferon |
US4897471A (en) | 1982-05-06 | 1990-01-30 | Amgen | Consensus human leukocyte interferon |
AU588819B2 (en) | 1984-10-29 | 1989-09-28 | Immunex Corporation | Cloning of human granulocyte-macrophage colony stimulating factor gene |
US5672502A (en) | 1985-06-28 | 1997-09-30 | Celltech Therapeutics Limited | Animal cell culture |
US4968607A (en) | 1987-11-25 | 1990-11-06 | Immunex Corporation | Interleukin-1 receptors |
US5075222A (en) | 1988-05-27 | 1991-12-24 | Synergen, Inc. | Interleukin-1 inhibitors |
EP0367566A1 (en) | 1988-10-31 | 1990-05-09 | Immunex Corporation | Interleukin-4 receptors |
US5856296A (en) | 1988-10-31 | 1999-01-05 | Immunex Corporation | DNA encoding interleukin-4 receptors |
US5395760A (en) | 1989-09-05 | 1995-03-07 | Immunex Corporation | DNA encoding tumor necrosis factor-α and -β receptors |
US5610279A (en) | 1989-09-12 | 1997-03-11 | Hoffman-La Roche Inc. | Human TNF receptor |
US6204363B1 (en) | 1989-10-16 | 2001-03-20 | Amgen Inc. | Stem cell factor |
US5272064A (en) | 1989-12-19 | 1993-12-21 | Amgen Inc. | DNA molecules encoding platelet-derived growth factor B chain analogs and method for expression thereof |
US5149792A (en) | 1989-12-19 | 1992-09-22 | Amgen Inc. | Platelet-derived growth factor B chain analogs |
EP0460846A1 (en) | 1990-06-05 | 1991-12-11 | Immunex Corporation | Type II interleukin-1 receptors |
US5767064A (en) | 1990-06-05 | 1998-06-16 | Immunex Corporation | Soluble type II interleukin-1 receptors and methods |
WO1994010308A1 (en) | 1992-10-23 | 1994-05-11 | Immunex Corporation | Methods of preparing soluble, oligomeric proteins |
WO1994028391A1 (en) | 1993-05-24 | 1994-12-08 | Immunex Corporation | Ligands for flt3 receptors |
US5981713A (en) | 1994-10-13 | 1999-11-09 | Applied Research Systems Ars Holding N.V. | Antibodies to intereleukin-1 antagonists |
WO1997001633A1 (en) | 1995-06-29 | 1997-01-16 | Immunex Corporation | Cytokine that induces apoptosis |
US6015938A (en) | 1995-12-22 | 2000-01-18 | Amgen Inc. | Osteoprotegerin |
US6096728A (en) | 1996-02-09 | 2000-08-01 | Amgen Inc. | Composition and method for treating inflammatory diseases |
US6271349B1 (en) | 1996-12-23 | 2001-08-07 | Immunex Corporation | Receptor activator of NF-κB |
US6235883B1 (en) | 1997-05-05 | 2001-05-22 | Abgenix, Inc. | Human monoclonal antibodies to epidermal growth factor receptor |
US6337072B1 (en) | 1998-04-03 | 2002-01-08 | Hyseq, Inc. | Interleukin-1 receptor antagonist and recombinant production thereof |
WO2001036637A1 (en) | 1999-11-17 | 2001-05-25 | Immunex Corporation | Receptor activator of nf-kappa b |
US6544424B1 (en) | 1999-12-03 | 2003-04-08 | Refined Technology Company | Fluid filtration system |
Non-Patent Citations (29)
Title |
---|
"Animal cell culture: A Practical Approach", 1992, OXFORD UNIVERSITY PRESS |
"Current Protocols in Molecular Biology", 1988, WILEY & SONS |
"Growth Factors: A Practical Approach", 1993, OXFORD UNIVERSITY PRESS INC. |
"Human Cytokines: Handbook for Basic and Clinical Research. all volumes", 1998, BLACKWELL SCIENCES |
"Leukocyte Typine VI", 1996 |
"Remington's Pharmaceutictll Sciences", 1995, MACK PUBLISHING COMPANY |
"The Cytokine Handbook", vol. 1, 2, ACADEMIC PRESS |
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1992, GREENE PUBLISHING ASSOCIATES |
BRASEL ET AL., BLOOD, vol. 88, 1996, pages 2004 - 2012 |
CHU; ROBINSON, CURRENT OPIN. BIOTECHNOL., vol. 12, 2001, pages 180 - 87 |
DO; CHEN-KIANG, CYTOKINE GROWTH FACTOR REV., vol. 13, no. 1, 2002, pages 19 - 25 |
FUREY, GEN. ENG. NEWS., vol. 22, no. 7, 2002, pages 62 - 63 |
HÅKANSSON ET AL., STRUCTURE, vol. 7, 1999, pages 255 - 64 |
HARBURY ET AL., NATURE, vol. 371, 1994, pages 80 - 83 |
HARBURY ET AL., SCIENCE, vol. 262, 1993, pages 1401 - 05 |
HARLOW; LANE: "Antibodies: A Laboratory Manual", 1990, COLD SPRING HARBOR LABORATORY PRESS, COLD SPRING HARBOR |
KAUFMAN ET AL., J. BIOL CHEM, vol. 263, 1988, pages 6352 - 6362 |
KAUFMAN R.J., METH ENZVMOL, vol. 185, 1990, pages 537 - 566 |
KAUFMAN, R.J., LARGE SCALE MAMMALIAN CELL CULTURE, 1990, pages 15 - 69 |
LOVEJOY ET AL., SCIENCE, vol. 259, 1993, pages 1288 - 1293 |
MAISONPIERRE ET AL., SCIENCE, vol. 277, no. 5322, 1997, pages 55 - 60 |
MCKINNON ET AL., J MOL ENDOCRINOL, vol. 6, 1991, pages 231 - 239 |
RUEGG; PYTELA, GENE, vol. 160, 1995, pages 257 - 62 |
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING LABORATORY PRESS |
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 2001, COLD SPRING HARBOR LABORATORY PRESS |
STETTLER, BIOTECHNOL BIOENG. DEC 20, vol. 95, no. 6, 2006, pages 1228 - 33 |
URLAUB ET AL., PROC NATL ACAD SCI USA, vol. 77, 1980, pages 4216 - 4220 |
VOISARD ET AL., BIOTECHNOLOGY AND BIOENGINEERING, vol. 82, 2003, pages 751 - 65 |
WOOD ET AL., J. IMMUNOL., vol. 145, 1990, pages 3011 - 3016 |
Cited By (121)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US10119118B2 (en) | 2006-09-13 | 2018-11-06 | Abbvie Inc. | Modified serum-free cell culture medium |
US8906646B2 (en) | 2006-09-13 | 2014-12-09 | Abbvie Inc. | Fed-batch method of making human anti-TNF-alpha antibody |
US8911964B2 (en) | 2006-09-13 | 2014-12-16 | Abbvie Inc. | Fed-batch method of making human anti-TNF-alpha antibody |
US9090867B2 (en) | 2006-09-13 | 2015-07-28 | Abbvie Inc. | Fed-batch method of making anti-TNF-alpha antibody |
US9234032B2 (en) | 2006-09-13 | 2016-01-12 | Abbvie Inc. | Fed-batch methods for producing adalimumab |
US9284371B2 (en) | 2006-09-13 | 2016-03-15 | Abbvie Inc. | Methods of producing adalimumab |
US9073988B2 (en) | 2006-09-13 | 2015-07-07 | Abbvie Inc. | Fed batch method of making anti-TNF-alpha antibodies |
US9109010B2 (en) | 2008-10-20 | 2015-08-18 | Abbvie Inc. | Viral inactivation during purification of antibodies cross reference to related applications |
US8895709B2 (en) | 2008-10-20 | 2014-11-25 | Abbvie Inc. | Isolation and purification of antibodies using protein A affinity chromatography |
US9018361B2 (en) | 2008-10-20 | 2015-04-28 | Abbvie Inc. | Isolation and purification of antibodies using protein a affinity chromatography |
US9090688B2 (en) | 2011-04-27 | 2015-07-28 | Abbvie Inc. | Methods for controlling the galactosylation profile of recombinantly-expressed proteins |
US9255143B2 (en) | 2011-04-27 | 2016-02-09 | Abbvie Inc. | Methods for controlling the galactosylation profile of recombinantly-expressed proteins |
US9062106B2 (en) | 2011-04-27 | 2015-06-23 | Abbvie Inc. | Methods for controlling the galactosylation profile of recombinantly-expressed proteins |
US9365645B1 (en) | 2011-04-27 | 2016-06-14 | Abbvie, Inc. | Methods for controlling the galactosylation profile of recombinantly-expressed proteins |
US9505834B2 (en) | 2011-04-27 | 2016-11-29 | Abbvie Inc. | Methods for controlling the galactosylation profile of recombinantly-expressed proteins |
US10213508B2 (en) | 2011-10-18 | 2019-02-26 | Coherus Biosciences, Inc. | Etanercept formulations stabilized with xylitol |
US10493151B2 (en) | 2011-10-18 | 2019-12-03 | Coherus Biosciences, Inc. | Etanercept formulations stabilized with sodium chloride |
US11000588B2 (en) | 2011-10-18 | 2021-05-11 | Coherus Biosciences, Inc. | Etanercept formulations stabilized with sodium chloride |
US11129876B2 (en) | 2011-10-18 | 2021-09-28 | Coherus Biosciences, Inc. | Etanercept formulations stabilized with amino acids |
US10293049B2 (en) | 2011-10-18 | 2019-05-21 | Coherus Biosciences, Inc. | Etanercept formulations stabilized with amino acids |
US10772963B2 (en) | 2011-10-18 | 2020-09-15 | Coherus Biosciences, Inc. | Etanercept formulations stabilized with xylitol |
US9181572B2 (en) | 2012-04-20 | 2015-11-10 | Abbvie, Inc. | Methods to modulate lysine variant distribution |
US9334319B2 (en) | 2012-04-20 | 2016-05-10 | Abbvie Inc. | Low acidic species compositions |
US9150645B2 (en) | 2012-04-20 | 2015-10-06 | Abbvie, Inc. | Cell culture methods to reduce acidic species |
US9505833B2 (en) | 2012-04-20 | 2016-11-29 | Abbvie Inc. | Human antibodies that bind human TNF-alpha and methods of preparing the same |
US9683033B2 (en) | 2012-04-20 | 2017-06-20 | Abbvie, Inc. | Cell culture methods to reduce acidic species |
US9193787B2 (en) | 2012-04-20 | 2015-11-24 | Abbvie Inc. | Human antibodies that bind human TNF-alpha and methods of preparing the same |
US9708400B2 (en) | 2012-04-20 | 2017-07-18 | Abbvie, Inc. | Methods to modulate lysine variant distribution |
US9359434B2 (en) | 2012-04-20 | 2016-06-07 | Abbvie, Inc. | Cell culture methods to reduce acidic species |
US9346879B2 (en) | 2012-04-20 | 2016-05-24 | Abbvie Inc. | Protein purification methods to reduce acidic species |
US9957318B2 (en) | 2012-04-20 | 2018-05-01 | Abbvie Inc. | Protein purification methods to reduce acidic species |
US9249182B2 (en) | 2012-05-24 | 2016-02-02 | Abbvie, Inc. | Purification of antibodies using hydrophobic interaction chromatography |
US10822429B2 (en) | 2012-07-09 | 2020-11-03 | Coherus Biosciences, Inc. | Etanercept formulations exhibiting marked reduction in sub-visible particles |
US9234033B2 (en) | 2012-09-02 | 2016-01-12 | Abbvie, Inc. | Methods to control protein heterogeneity |
US9290568B2 (en) | 2012-09-02 | 2016-03-22 | Abbvie, Inc. | Methods to control protein heterogeneity |
US9512214B2 (en) | 2012-09-02 | 2016-12-06 | Abbvie, Inc. | Methods to control protein heterogeneity |
US9206390B2 (en) | 2012-09-02 | 2015-12-08 | Abbvie, Inc. | Methods to control protein heterogeneity |
US10954293B2 (en) | 2012-09-11 | 2021-03-23 | Coherus Biosciences, Inc. | Correctly folded etanercept in high purity and excellent yield |
US10954295B2 (en) | 2012-09-11 | 2021-03-23 | Coherus Biosciences, Inc. | Correctly folded etanercept in high purity and excellent yield |
US10954294B2 (en) | 2012-09-11 | 2021-03-23 | Coherus Biosciences, Inc. | Correctly folded etanercept in high purity and excellent yield |
US11001627B2 (en) | 2012-09-11 | 2021-05-11 | Coherus Biosciences, Inc. | Correctly folded etanercept in high purity and excellent yield |
US10947306B2 (en) | 2012-09-11 | 2021-03-16 | Coherus Biosciences, Inc. | Correctly folded etanercept in high purity and excellent yield |
WO2014109858A1 (en) * | 2013-01-14 | 2014-07-17 | Amgen Inc. | Methods of using cell-cycle inhibitors to modulate one or more properties of a cell culture |
TWI625390B (en) * | 2013-03-14 | 2018-06-01 | 安美基公司 | Methods for increasing mannose content of recombinant proteins |
US9067990B2 (en) | 2013-03-14 | 2015-06-30 | Abbvie, Inc. | Protein purification using displacement chromatography |
US11952605B2 (en) | 2013-03-14 | 2024-04-09 | Amgen Inc. | Methods for increasing mannose content of recombinant proteins |
EP3434760A1 (en) | 2013-03-14 | 2019-01-30 | Amgen, Inc | Methods for increasing mannose content of recombinant proteins |
US9499614B2 (en) | 2013-03-14 | 2016-11-22 | Abbvie Inc. | Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides |
US8921526B2 (en) | 2013-03-14 | 2014-12-30 | Abbvie, Inc. | Mutated anti-TNFα antibodies and methods of their use |
EP4141104A1 (en) | 2013-03-14 | 2023-03-01 | Amgen Inc. | Methods for increasing mannose content of recombinant proteins |
US9822388B2 (en) | 2013-03-14 | 2017-11-21 | Amgen Inc. | Methods for increasing mannose content of recombinant proteins |
WO2014159259A1 (en) | 2013-03-14 | 2014-10-02 | Amgen Inc. | Methods for increasing mannose content of recombinant proteins |
US11459595B2 (en) | 2013-03-14 | 2022-10-04 | Amgen Inc. | Methods for increasing mannose content of recombinant proteins |
US9708399B2 (en) | 2013-03-14 | 2017-07-18 | Abbvie, Inc. | Protein purification using displacement chromatography |
US11319568B2 (en) | 2013-03-14 | 2022-05-03 | Amgen Inc. | Methods for increasing mannose content of recombinant proteins |
AU2014241259B9 (en) * | 2013-03-26 | 2018-12-20 | Coherus Biosciences, Inc. | Protein production method |
US10975138B2 (en) | 2013-03-26 | 2021-04-13 | Coherus Biosciences, Inc. | Correctly folded etanercept composition |
EP2978498A4 (en) * | 2013-03-26 | 2016-10-05 | Coherus Biosciences Inc | Protein production method |
AU2014241259B2 (en) * | 2013-03-26 | 2018-12-06 | Coherus Biosciences, Inc. | Protein production method |
EP3391942A1 (en) * | 2013-03-26 | 2018-10-24 | Coherus Biosciences, Inc. | Protein production method |
JP2016514475A (en) * | 2013-03-26 | 2016-05-23 | コヒラス・バイオサイエンシズ・インコーポレイテッド | Protein production method |
AU2019201129B2 (en) * | 2013-03-26 | 2020-12-24 | Coherus Biosciences, Inc. | Protein production method |
US10421987B2 (en) | 2013-05-30 | 2019-09-24 | Amgen Inc. | Methods for increasing mannose content of recombinant proteins |
US11597959B2 (en) | 2013-05-30 | 2023-03-07 | Amgen Inc. | Methods for increasing mannose content of recombinant proteins |
US10894972B2 (en) | 2013-05-30 | 2021-01-19 | Amgen Inc. | Methods for increasing mannose content of recombinant proteins |
US11434514B2 (en) | 2013-05-30 | 2022-09-06 | Amgen Inc. | Methods for increasing mannose content of recombinant proteins |
US9481901B2 (en) | 2013-05-30 | 2016-11-01 | Amgen Inc. | Methods for increasing mannose content of recombinant proteins |
US10184143B2 (en) | 2013-05-30 | 2019-01-22 | Amgen Inc. | Methods for increasing mannose content of recombinant proteins |
US11946085B2 (en) | 2013-05-30 | 2024-04-02 | Amgen Inc. | Methods for increasing mannose content of recombinant proteins |
US9598667B2 (en) | 2013-10-04 | 2017-03-21 | Abbvie Inc. | Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins |
US11390663B2 (en) | 2013-10-11 | 2022-07-19 | Regeneron Pharmaceuticals, Inc. | Metabolically optimized cell culture |
US9085618B2 (en) | 2013-10-18 | 2015-07-21 | Abbvie, Inc. | Low acidic species compositions and methods for producing and using the same |
US9266949B2 (en) | 2013-10-18 | 2016-02-23 | Abbvie, Inc. | Low acidic species compositions and methods for producing and using the same |
US9499616B2 (en) | 2013-10-18 | 2016-11-22 | Abbvie Inc. | Modulated lysine variant species compositions and methods for producing and using the same |
US9200069B2 (en) | 2013-10-18 | 2015-12-01 | Abbvie, Inc. | Low acidic species compositions and methods for producing and using the same |
US9522953B2 (en) | 2013-10-18 | 2016-12-20 | Abbvie, Inc. | Low acidic species compositions and methods for producing and using the same |
US9200070B2 (en) | 2013-10-18 | 2015-12-01 | Abbvie, Inc. | Low acidic species compositions and methods for producing and using the same |
US9688752B2 (en) | 2013-10-18 | 2017-06-27 | Abbvie Inc. | Low acidic species compositions and methods for producing and using the same using displacement chromatography |
US8946395B1 (en) | 2013-10-18 | 2015-02-03 | Abbvie Inc. | Purification of proteins using hydrophobic interaction chromatography |
US9181337B2 (en) | 2013-10-18 | 2015-11-10 | Abbvie, Inc. | Modulated lysine variant species compositions and methods for producing and using the same |
US9315574B2 (en) | 2013-10-18 | 2016-04-19 | Abbvie, Inc. | Low acidic species compositions and methods for producing and using the same |
US9017687B1 (en) | 2013-10-18 | 2015-04-28 | Abbvie, Inc. | Low acidic species compositions and methods for producing and using the same using displacement chromatography |
JP2016534732A (en) * | 2013-10-31 | 2016-11-10 | アムジエン・インコーポレーテツド | Use of monensin to regulate glycosylation of recombinant proteins |
US9550826B2 (en) | 2013-11-15 | 2017-01-24 | Abbvie Inc. | Glycoengineered binding protein compositions |
WO2015095809A1 (en) | 2013-12-20 | 2015-06-25 | Biogen Idec Ma Inc. | Use of perfusion seed cultures to improve biopharmaceutical fed-batch production capacity and product quality |
WO2015105609A1 (en) | 2014-01-13 | 2015-07-16 | Amgen Inc. | Regulating ornithine metabolism to manipulate the high mannose glycoform content of recombinant proteins |
US11254963B2 (en) | 2014-01-13 | 2022-02-22 | Amgen Inc. | Increasing ornithine accumulation to increase high mannose glycoform content of recombinant proteins |
US10513723B2 (en) | 2014-01-13 | 2019-12-24 | Amgen Inc. | Decreasing ornithine production to decrease high mannose glycoform content of recombinant proteins |
US11001623B2 (en) | 2014-01-30 | 2021-05-11 | Coherus Biosciences, Inc. | Method of manufacturing a protein by perfusion in media with a low amino acid concentration |
US10047141B2 (en) | 2014-01-30 | 2018-08-14 | Coherus Biosciences, Inc. | Method of manufacturing a protein by perfusion in media with a low amino acid concentration |
EP4372078A2 (en) | 2014-06-04 | 2024-05-22 | Amgen Inc. | Methods for harvesting mammalian cell cultures |
JP2020124207A (en) * | 2014-06-04 | 2020-08-20 | アムジエン・インコーポレーテツド | Methods for harvesting mammalian cell cultures |
JP7377316B2 (en) | 2014-06-04 | 2023-11-09 | アムジエン・インコーポレーテツド | Methods for harvesting mammalian cell cultures |
WO2015188009A1 (en) | 2014-06-04 | 2015-12-10 | Amgen Inc. | Methods for harvesting mammalian cell cultures |
JP2022126811A (en) * | 2014-06-04 | 2022-08-30 | アムジエン・インコーポレーテツド | Methods for harvesting mammalian cell cultures |
EP3926051A1 (en) | 2014-06-04 | 2021-12-22 | Amgen, Inc | Methods for harvesting mammalian cell cultures |
JP7097404B2 (en) | 2014-06-04 | 2022-07-07 | アムジエン・インコーポレーテツド | Methods for Retrieving Mammalian Cell Cultures |
US11427848B2 (en) | 2014-06-04 | 2022-08-30 | Amgen Inc. | Methods for harvesting mammalian cell cultures |
JP2017519497A (en) * | 2014-06-04 | 2017-07-20 | アムジエン・インコーポレーテツド | Method for recovering mammalian cell culture |
US11384378B2 (en) | 2014-06-04 | 2022-07-12 | Amgen Inc. | Methods for harvesting mammalian cell cultures |
US11060058B2 (en) | 2014-06-06 | 2021-07-13 | Genzyme Corporation | Perfusion culturing methods and uses thereof |
US12006510B2 (en) | 2014-06-06 | 2024-06-11 | Genzyme Corporation | Perfusion culturing methods and uses thereof |
EP3152299B1 (en) | 2014-06-09 | 2019-11-20 | Genzyme Corporation | Seed train processes and uses thereof |
JP2017521057A (en) * | 2014-06-09 | 2017-08-03 | ジェンザイム・コーポレーション | Seed train method and its use |
WO2016089919A1 (en) | 2014-12-01 | 2016-06-09 | Amgen Inc. | Process for manipulating the level of glycan content of a glycoprotein |
US10167492B2 (en) | 2014-12-01 | 2019-01-01 | Amgen Inc. | Process for manipulating the level of glycan content of a glycoprotein |
US10822630B2 (en) | 2014-12-01 | 2020-11-03 | Amgen Inc. | Process for manipulating the level of glycan content of a glycoprotein |
EP3680344A1 (en) | 2014-12-01 | 2020-07-15 | Amgen Inc. | Process for manipulating the level of glycan content of a glycoprotein |
EP3227454B1 (en) | 2014-12-01 | 2020-01-29 | Amgen Inc. | Process for manipulating the level of glycan content of a glycoprotein |
US10131891B2 (en) | 2014-12-31 | 2018-11-20 | Lg Chem, Ltd. | Method of using insulin for controlling glycosylation of recombinant glycoprotein |
JP2018500041A (en) * | 2014-12-31 | 2018-01-11 | エルジー・ケム・リミテッド | Methods for regulating glycosylation of recombinant glycoproteins |
IL261411B1 (en) * | 2016-03-02 | 2023-11-01 | Lonza Ag | Improved fermentation process |
IL261411B2 (en) * | 2016-03-02 | 2024-03-01 | Lonza Ag | Improved fermentation process |
WO2017149065A1 (en) | 2016-03-02 | 2017-09-08 | Lonza Ltd | Improved fermentation process |
US11377677B2 (en) | 2016-03-02 | 2022-07-05 | Lonza Ltd | Fermentation process |
US11702628B2 (en) | 2017-03-31 | 2023-07-18 | Boehringer Ingelheim International Gmbh | Perfusion medium for culturing mammalian cells and reducing cell bleed in a perfusion cell culture |
US12084686B2 (en) | 2018-05-01 | 2024-09-10 | Amgen Inc. | Antibodies with modulated glycan profiles |
WO2022036275A3 (en) * | 2020-08-14 | 2022-05-12 | Bristol-Myers Squibb Company | Manufacturing process for protein |
WO2022047108A1 (en) * | 2020-08-31 | 2022-03-03 | Regeneron Pharmaceuticals, Inc. | Asparagine feed strategies to improve cell culture performance and mitigate asparagine sequence variants |
WO2024054414A1 (en) | 2022-09-06 | 2024-03-14 | Amgen Inc. | Lean perfusion cell culture methods |
WO2024059235A2 (en) | 2022-09-16 | 2024-03-21 | Amgen Inc. | A method for harvesting products from perfusion cultures |
Also Published As
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2020281075B2 (en) | Mammalian cell culture | |
WO2014159259A1 (en) | Methods for increasing mannose content of recombinant proteins |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 12738674 Country of ref document: EP Kind code of ref document: A2 |
|
ENP | Entry into the national phase |
Ref document number: 2838695 Country of ref document: CA |
|
WWE | Wipo information: entry into national phase |
Ref document number: MX/A/2013/014937 Country of ref document: MX |
|
ENP | Entry into the national phase |
Ref document number: 2014519184 Country of ref document: JP Kind code of ref document: A |
|
WWE | Wipo information: entry into national phase |
Ref document number: 201391826 Country of ref document: EA |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2013003792 Country of ref document: CL |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
ENP | Entry into the national phase |
Ref document number: 2012279230 Country of ref document: AU Date of ref document: 20120629 Kind code of ref document: A |
|
ENP | Entry into the national phase |
Ref document number: 20147001985 Country of ref document: KR Kind code of ref document: A |
|
REEP | Request for entry into the european phase |
Ref document number: 2012738674 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2012738674 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 14127050 Country of ref document: US |
|
REG | Reference to national code |
Ref country code: BR Ref legal event code: B01A Ref document number: 112013033730 Country of ref document: BR |
|
ENP | Entry into the national phase |
Ref document number: 112013033730 Country of ref document: BR Kind code of ref document: A2 Effective date: 20131227 |